SlideShare a Scribd company logo
www.thelancet.com/infection Vol 15 January 2015 95
Review
The effect of HIV infection on the host response to
bacterial sepsis
Michaëla A M Huson, Martin P Grobusch, Tom van der Poll
Bacterial sepsis is an important cause of morbidity and mortality in patients with HIV. HIV causes increased
susceptibility to invasive infections and affects sepsis pathogenesis caused by pre-existing activation and exhaustion
of the immune system. We review the effect of HIV on different components of immune responses implicated in
bacterial sepsis, and possible mechanisms underlying the increased risk of invasive bacterial infections. We focus
on pattern recognition receptors and innate cellular responses, cytokines, lymphocytes, coagulation, and the
complement system. A combination of factors causes increased susceptibility to infection and can contribute to a
disturbed immune response during a septic event in patients with HIV. HIV-induced perturbations of the immune
system depend on stage of infection and are only in part restored by combination antiretroviral therapy. Immuno-
modulatory treatments currently under development for sepsis might be particularly beneficial to patients with
HIV co-infection because many pathogenic mechanisms in HIV and sepsis overlap.
Introduction
People infected with HIV are at increased risk of
developing other infections. Reports from developed
countries show that, with the introduction of
combination antiretroviral therapy (cART), the incidence
of opportunistic infections such as Pneumocystis jirovecii
pneumonia has decreased substantially, and sepsis is
now the principal cause of intensive care unit (ICU)
admission and death in patients with HIV who are
admitted to hospital.1
Worldwide, patients with HIV are
at increased risk of developing bacterial bloodstream
infections, particularly with non-typhoidal salmonella
(NTS) and Streptococcus pneumoniae,2,3
even in those
using effective cART.4
In developing regions with high
rates of HIV infection, the scale of the problem is
immense. In African hospitals, 31–83% of patients
presenting with fever are infected with HIV, and 10–32%
of these patients have bacterial bloodstream infections,
with mortality rates of 25–46% (table 1).5-15
However, little
is known about the particularities of the host response
during bacterial sepsis in patients with HIV. The
immune response during sepsis is thought to be an
imbalance between proinflammatory and anti-
inflammatory effects, which result in organ failure.16,17
Likewise, HIV infection is characterised by a
combination of immune suppression and chronic
inflammation, which results in exhaustion of the
immune system.18
In this Review we describe how
different components of the immune response might be
affected by HIV during bacterial sepsis, and possible
mechanisms for the increased risk of invasive bacterial
infections. We also discuss parallels in immuno-
modulatory therapies proposed for sepsis and HIV,
which represent an appealing area for future research.
First line defences
Overview
A sepsis event typically starts with a pathogen invading
a normally sterile site. The first line of defence consists
of epithelial cells of the skin, gut, and lungs. After
breaching the epithelial barrier, pathogens are first
detected by innate immune cells, including monocytes,
macrophages, dendritic cells, and neutrophils.19
These
cells express pattern recognition receptors (PRRs) on
their surface and in the cytosol to enable recognition of
conserved motifs expressed by pathogens.20,21
Four
classes of PRRs have been identified: toll-like receptors
(TLRs), C-type lectin receptors (CLRs), nucleotide-
binding oligomerisation domain leucine-rich-repeat
containing receptors (NOD-LRRs), and retinoic acid-
inducible gene I protein helicase receptors (RLRs).20
In
the context of sepsis pathogenesis, TLRs have been
studied the most extensively. They are expressed on the
cell surface (TLRs 1, 2, 4, 5, and 6) to enable recognition
of bacterial outer membrane components, and in
intracellular compartments (TLRs 3, 7, 8, and 9) for
detection of nucleic acids derived from intracellular
pathogens, mainly viruses.20
For bacterial recognition,
TLRs 2, 4, 5, and 9 are the most important. Additionally,
TLR7 can sense bacterial RNA released within
phagosomal vacuoles,22
and TLR3 can function as an
amplifier of host RNA-triggered inflammation during
sepsis.23
Ligand recognition by TLRs triggers a
signalling cascade, which results in the production of
cytokines.20
Although essential for pathogen recognition
and the innate immune response, uncontrolled
stimulation causes excessive inflammation; hence TLR
signalling is normally tightly regulated.
Innate immune cells have several specific functions in
antibacterial immunity. Monocytes and macrophages
have a crucial role in maintaining homoeostasis by
phagocytosis of apoptotic cells and microorganisms.19
As the main producers of proinflammatory cytokines,
they are also thought to be key in sepsis pathogenesis.19
Dendritic cells are the main antigen-presenting cells;
maturation is induced after ingestion of antigen,
followed by migration to lymphoid tissue. Mature
dendritic cells express co-stimulatory molecules on
their surface, which synergise with antigen to activate
T cells.24
Natural killer (NK) cells are implicated in
Lancet Infect Dis 2015;
15: 95–108
Published Online
October 20, 2014
http://dx.doi.org/10.1016/
S1473-3099(14)70917-X
Division of Infectious Diseases,
Centre of Experimental and
Molecular Medicine
(M A M Huson MD,
ProfT van der Poll MD); and
Division of Infectious Diseases,
Centre ofTropical Medicine and
Travel Medicine, Academic
Medical Centre, University of
Amsterdam, Amsterdam,
Netherlands
(Prof M P Grobusch MD)
Correspondence to:
Dr Michaëla A M Huson,
Academic Medical Centre,
University of Amsterdam,
Amsterdam 1105 AZ,
Netherlands
m.a.huson@amc.uva.nl
96 www.thelancet.com/infection Vol 15 January 2015
Review
antibacterial immune responses through the ability to
directly lyse infected cells, provide early sources of
proinflammatory cytokines, predominantly inter-
feron-γ, induce dendritic cell maturation, and amplify
the proinflammatory effects of myeloid cells.25,26
Likewise, natural killer T cells (NK T cells), a subset that
shares cell-surface proteins with conventional T cells
and NK cells, have been implicated in sepsis
pathogenesis because of their strong proinflammatory
cytokine release.27
Recruited neutrophils form an
additional important first line of defence against
invading pathogens. They kill microbes through
phagocytosis, the release of lytic enzymes from their
granules, the production of reactive oxygen inter-
mediates, and the formation of neutrophil extracellular
traps (NETs)—lattices of chromatin decorated with anti-
microbial proteins with strong bactericidal capacity.28
The innate immune system is thought to be mostly
responsible for the excessive release of proinflammatory
cytokines early in sepsis pathogenesis.29
The most
extensively studied proinflammatory cytokines in
sepsis are tumour necrosis factor α (TNFα) and
interleukin 1β, both of which are capable of activating
target cells and induce the production of more
inflammatory mediators.29
Additionally, host cells
release damage-associated molecular patterns (DAMPs)
in response to pathogens or injury, which are recognised
by PRRs, thereby enhancing immune activation. The
most investigated DAMP is HMGB1, which signals via
TLR2, TLR4, and TLR9 to induce cytokine release,
activation of coagulation, and neutrophil recruitment.30
Proinflammatory cytokines and DAMPs seem to have a
double role in sepsis pathogenesis; they are essential
for an adequate innate defence during early stage
infection, but also contribute to hyperinflammation
during late phase uncontrolled infection.20,21
In patients
with severe sepsis and in murine sepsis models, high
concentrations of interleukins 1, 6, 8, and 17, CCL2,
CSF3, and HMGB1 are associated with mortality.29
HIV affects the first lines of defence in several ways.
First, defects of epithelial barriers are common and
have been particularly well described for the gut. In the
gut, HIV causes barrier defects during acute infection,
which are maintained during chronic infection, thereby
enabling invasive infections by intestinal pathogens
such as non-typhoidal salmonella.31
Furthermore,
microbial products that translocate into the circulation
fuel chronic immune activation and exhaustion.32
Toll-like receptors
HIV infection and AIDS are associated with increased
TLR2, TLR3, TLR4, TLR7, and TLR9 expression on
various cells, including T lymphocytes, monocytes,
macrophages, and dendritic cells,33–38
although one study34
reported lower peripheral blood mononuclear cell
(PBMC) RNA expression levels of TLR3, TLR4, and TLR9
in patients with chronic HIV unresponsive to cART.
Reports on the functional effect of differential TLR
expression are inconsistent. Ex-vivo studies with PBMCs
from patients with HIV noted a correlation between
increased expression of TLRs and increased TNFα
production after stimulation with lipopolysaccharide, a
Study location
andtimeframe
Primary
inclusion
criteria
Patients
infectedwith
HIV
(%ofpatients
tested)
CA
bacterial
BSI in
patients
with HIV
(%)
Main isolates in
patientswith
HIV (%)
Mortality in
patients
with HIV
withCA
bacterial BSI
Archibald,
19985
UrbanTanzania,
1995
Febrile
(≥37·5°C)
admission
282 (55%) 51 (18%) NTS (45%),
Escherichiacoli
(14%),
Streptococcus
pneumoniae
(12%)
Not reported
Archibald,
20006
Urban Malawi,
1997
Febrile
(≥37·5°C)
admissions
173 (74%) 37 (21%) Spneumoniae
(57%), NTS
(30%)
Not reported
Arthur,
20017
Urban Kenya,
1988–97
Hospital
admission
436 (32%) 87 (20%) NTS (46%),
Spneumoniae
(33%), Ecoli (6%)
39%
Bell, 20018
Urban Malawi,
1998
Febrile
(37·5°C)
admissions
173 (73%) 36 (21%) NTS (62%), Ecoli
(7%), Salmonella
Typhi (7%)
Not reported
Crump,
20119
RuralTanzania,
2007–08
Febrile
(≥38°C)
admissions
161 (40%) 26 (16%) Spneumoniae
(54%), Ecoli
(12%), NTS (8%),
STyphi (8%)
Not reported
Grant,
199710
Ivory coast,
1995
Admission
tothe
infectious
diseaseunit
198 (79%) 39 (20%) NTS (59%), Ecoli
(15%),
Spneumoniae
(10%)
46%
Mayanja,
201011
RuralUganda,
1996–2007
Fever
(≥38°C)with
no
detectable
malaria
parasites
488 (64%) 152 (31%) Spneumoniae
(43%), NTS
(26%), Ecoli (6%)
Not reported
Meremo,
201212
UrbanTanzania,
2011
Hospital
admission
with fever
(>37·5°C)
156 (45%) 16 (10%) NTS (75%) 35%
Nadjm,
201213
RuralTanzania,
2007
Fever and
one severity
criterion
69 (35%) 12 (17%) NTS (25%),
Spneumoniae
(25%),
Streptococcus
pyogenes (17%)
25%
Peters,
200414
Urban Malawi,
2000
Admission
with fever
(≥37·4°C)or
historyof
fever inthe
past 4days
291 (83%) 66 (23%) NTS (67%),
Spneumoniae
(20%), Ecoli (6%)
Not reported
Ssali,
199815
Uganda, 1997 Admission
with fever
(>38°C)
222 (76%) 33 (15%) Salmonella spp
(39%),
Spneumoniae
(33%)
Not reported
Data was derived from prospective observational studies, which were previously described by Huson and colleagues.3
CA=community acquired. BSI=bloodstream infection. NTS=non-typhoidal salmonella.
Table 1: Burden of community-acquired bacterial bloodstream infections in patients with HIV in African
countries where HIV is highly prevalent
www.thelancet.com/infection Vol 15 January 2015 97
Review
See Online for appendix
TLR4 ligand.36,38
By contrast, alveolar macrophages from
patients with HIV showed a classic activation phenotype
with increased TLR4 expression, although binding,
internalisation, and killing of opsonised S pneumoniae
bacteria, which signal via TLR2, TLR4, and TLR9, were
similar to HIV-negative controls.37
TLR3 and TLR7 are
known to be key modulators in anti-HIV immunity, and
HIV-induced changes in expression of these TLRs might
have a role during antibacterial defence, but no studies
have yet investigated this subject.
Most evidence suggests that upregulation of TLRs is
responsible for cell activation in response to bacteria in
patients with HIV, especially in those with advanced
disease, although functional consequences differ
according to cell type. Increased expression of TLRs on
cytokine-producing cells might contribute to aberrant
inflammation in the disturbed homoeostasis in sepsis,
although this possibility has not yet been addressed in
in-vivo studies.
Monocytes and macrophages
The effect of HIV on monocytes and macrophages is
mainly indirect because only a small percentage of
these cells are infected with HIV.39
HIV-mediated
defects in phagocytosis, cell signalling, and cytokine
production have been described, although results vary
between studies (appendix).
Ex-vivo studies using monocytes or monocyte-derived
macrophages from patients with HIV, reported reduced
phagocytosis of Escherichia coli40
and, in symptomatic
patients with HIV, Staphylococcus aureus.41
By contrast,
others observed normal phagocytic function of
macrophages towards opsonised E coli and S aureus in
vitro,42
and increased monocyte phagocytosis of these
pathogens in cells harvested from patients with early
HIV infection.43
In peripheral blood, both monocytes
and macrophages appear to show an HIV-induced,
primed, proinflammatory state, with increased cytokine
release on stimulation with TLR ligands.44,45
A non-classic
subset of monocytes with high expression of M-DC8, a
subset that is also increased in inflamed tissue in chronic
inflammatory diseases such as rheumatoid arthritis, was
identified as the predominant cell type responsible for
TNFα overproduction46
(table 2).47–67
Several studies also examined the effect of HIV on the
alveolar compartment. Although normal alveolar
Study condition* HIV-induced changes Comments
Whole blood In vivo ↑ Interleukin 6, interleukin 10,TNFα,
interleukin 1α, interleukin 1β, interleukin 8,
CCL2, interleukin 1R1, and interferon γ47,48
↓ interleukin 12, andCSF248,49
Cytokine levels normalised aftertreatmentwith cART47,48
Two studies50,51
examined cytokine levels in sepsis patients, but recorded no
differences, except higher interleukin 10 concentrations in HIV co-infected
patients inoneofthese studies50
DecreasedCSF2 levelswere seen in patientswith HIVwith advanceddisease
Peripheral blood
mononuclear
cells
Ex-vivo and in-
vitro stimulation
↑TNFα 38,46,52
··
Monocytes Ex-vivo
stimulation
↑ Interleukin 6, interleukin 1β,TNFα44
··
M-DC8
monocytes
Ex-vivo
stimulation
↑TNFα46
These cellswere identified asthe main celltype responsible forTNFα
overproduction46
Increased levelsof M-DC8 monocyteswere recorded in viraemic patients
with HIV, but not in virally suppressed patientson cART46
Alveolar
macrophages
Ex-vivo
stimulation
↓TNFα, interleukin 8, interleukin 1253–56
Reports conflictonthis subject;other studies noted increased interleukin 8,
12 and 10 in alveolar macrophagesof patientswith HIV56
Impaired interleukin 12 productionwasonly reported in patientswith
advanceddisease56
Dendritic cells In-vitro and ex-
vivo stimulation
↓ Interleukin 12,57–59
interleukin 6,58
interferon γ,γ,58,6058,60
interferon α,interferon α,59,6159,61
interleukin 2,interleukin 2,6161
and interleukin 10and interleukin 106060
Most reportsdescribedecreased cytokine production after stimulation, but
normal,62
or even increased cytokine responsesto stimulationwere also
reported63
Natural killer
cells
Ex-vivo
stimulation
↓ Interferon γ64
The effectwasobserved in both cART-treated anduntreated patients64
Natural killer
T cells
Ex-vivo
stimulation
↓ Interferon γ and interleukin 465
Stimulationsweredonewith marine sponge-derived α-galactosylceramide
(αGalCer) as a ligand;65
similar studiesusing bacterial ligands as a stimulus
were not available
Cytokine secretionwas restored by cART65
CD4T cells In-vivo ↑ Interleukin 1066
Interleukin 10 productionwas increased in chronic progressors and recently
infected individuals, but not in non-progressorswith HIV66
Gut mucosa
Th17 cells
Ex-vivo
stimulation
↓ Interleukin 22,TNFα, and interferon γ67
↑ Interleukin 1067
Restorationof cytokine responseswas seen after prolongedtreatmentwith
cART67
cART=combination antiretroviral therapy. *All studies are in human beings or used human cells, or human cell lines; all stimulations were done with bacteria or bacterial
products unless otherwise specified.
Table 2: Effect of HIV infection on the cytokine milieu by cell type
98 www.thelancet.com/infection Vol 15 January 2015
Review
macrophage phagocytosis of S pneumoniae was
previously reported,37
a study68
noted impaired phago-
cytosis in HIV-infected small alveolar macrophages, as
well as impaired proteolysis of phagosomes in both
infected and uninfected alveolar macrophages.
Cytokine release, including that of interleukin 8, in
response to S pneumoniae, and TNFα, in response to
TLR2 and TLR4 ligands, was impaired in alveolar
macrophages from patients with HIV (table 2).53–55
Considering the crucial role of TNFα in resisting
experimental S pneumoniae infections in mice,69
and
the established part in neutrophil recruitment to sites
of infection by interleukin 8, a reduced cytokine
response by alveolar macrophages might contribute to
increased susceptibility to invasive pneumococcal
disease in patients with HIV (figure 1). Another study56
noted increased production of both proinflammatory
(interleukin 8 and 12) and anti-inflammatory
(interleukin 10) cytokines by alveolar macrophages of
asymptomatic patients with HIV after stimulation with
S aureus. However, in patients with more advanced
disease interleukin 12 release was reduced after
stimulation, which was mediated by interleukin 10.56
Interleukin 12 is known to induce T-helper (Th) 1
development, which is essential for defence against
intracellular pathogens, such as non-typhoidal
salmonella, but also has a role in the immune response
against S pneumoniae.70
Finally, alveolar macrophage
apoptosis, a critical host cell response for control of
infection, including pneumococcal pneumonia,71
might
be impaired in patients with HIV.72
Most evidence suggests impairment of phagocytosis
by macrophages in individuals with HIV, which is
accompanied by impaired proinflammatory cytokine
release, especially in alveolar macrophages. These
defects might contribute to increased susceptibility to
bacterial infection and deregulated inflammation.
Dendritic cells
In patients with HIV, around 10–15% of dendritic cells
are infected,57
but HIV also affects dendritic cell
function in indirect ways (appendix). Patients with HIV
have reduced peripheral blood dendritic cell
numbers,61,63,73,74
which are inversely correlated to viral
↑S pneumoniae
↓Interleukin 17
↓CD4
T cells ↓Macrophage recruitment
↓Macrophage phagocytosis
↓TNFα
↓Interleukin 8
↓Neutrophil recruitment
Bloodstream invasion of S pneumoniae
Alveolar interstitium
Nasopharyngeal cavity
↓Opsonic capacity of IgA ( ) and IgG ( )
Bloodstream
Alveolar space
Nasopharynx interstitium
Figure 1: Alterations in the host response of patient with HIV that might cause increased susceptibility to invasive infection with Streptococcus pneumoniae
In the upper airways, impaired recruitment of macrophages due to CD4 T-cell depletion and low concentrations of interleukin 17 enable colonisation by
S pneumoniae on the respiratory epithelium. In the lower airways, impaired antipneumococcal activity of IgA and IgG on the mucosal surface of the respiratory
epithelium allows the attachment of S pneumoniae to the epithelial cells and migration of the pathogen across the epithelial barrier. Ineffective opsonisation also
impairs phagocytosis by alveolar macrophages. Furthermore, alveolar macrophages from HIV-positive patients showed decreased TNFα production in response
to TLR2 and TLR4 ligands, and concentrations of TNFα in bronchoalveolar lavage fluid are reduced in patients with HIV, thereby hindering an effective
proinflammatory response. Alveolar macrophages from patients with HIV produce lower concentrations of interleukin 8 in response to S pneumoniae, resulting
in less effective recruitment of neutrophils. Neutrophils of HIV patients are also less able to respond to chemotactic signals, such as interleukin 8, but also
pneumolysin derived from the bacterium, further impairing a massive neutrophil influx. The combination of impairments in antipneumococcal defence enables
the invasion of S pneumoniae into the bloodstream. TNFα=tumour necrosis factor α.
www.thelancet.com/infection Vol 15 January 2015 99
Review
load,74
and positively correlated with CD4 counts.61
Homing of dendritic cells in lymph nodes75
and
increased apoptosis76
are possible explanations.
Reports on the effect of HIV on dendritic cell
maturation are inconsistent. An atypical phenotype of
immune activation without full maturation has been
reported,56,77
as well as reduced capacity to mature in
response to stimuli combined with interleukin 10
induction and immune suppression.78
By contrast,
normal maturation in response to stimuli57,73
and HIV-
induced maturation have also been described.79
Stimulation-induced production of cytokines,
including interleukin 12,57–59
interleukin 6,58
interferon
γ,58,59
interferon α,59,61
interleukin 2,60
and interleukin
10,60
was impaired by HIV, as well as dendritic cell-
mediated activation of T cells (table 2).79
Reduced
interleukin 12 production was associated with impaired
activation of NK cells in vitro.59
Some studies, however,
reported normal62
or even increased cytokine responses
of dendritic cells to stimuli.63
The number of dendritic cells in peripheral blood is
reduced in patients with HIV, and, additionally, most
studies report impaired dendritic cell function. HIV-
induced tolerance to antigen and reduced maturation
of dendritic cells might compromise immunity against
various of pathogens, rendering patients with HIV
more susceptible to sepsis.
NK cells and NK T cells
Reduced NK cell numbers and function during HIV
infection contribute to decreased resistance against
HIV and other pathogens (appendix).80,81
Ex-vivo
stimulation of NK cells from patients infected with HIV
with E coli or Salmonella typhimurium showed that both
untreated and treated patients had lower overall
frequencies of responsive interferon-γ-producing NK
cells (table 2).64
Although this study is the only one to
specifically address antibacterial immune responses by
NK cells from patients with HIV, many studies report
on HIV-induced changes that prevent an effective anti-
HIV NK cell response, which might also result in
impaired antibacterial defence. First, correlating with
viral load, HIV infection induces expansion of a CD56-
negative NK cell subpopulation, which is unresponsive
to stimulation and is thought to represent an exhausted
state, with concomitant loss of responsive CD56-
positive NK cell subsets due to increased apoptosis.80,82
CD56-negative NK cells are also defective in their
interaction with dendritic cells, thereby contributing to
the accumulation of immature dendritic cells.80,83
Second, HIV causes shedding of MHC class I chain-
related molecules MICA and MICB from the surface of
infected cells. By binding to the NK cell receptor
NKG2D, soluble MICA and MICB provide a negative
feedback signal, resulting in subsequent down-
regulation of NKG2D, thereby promoting the
generation of anergic NK cells.82
Third, decreased
production of chemokines impairs chemotactic
signalling to other immune cells.80,84
NK cells from
patients with HIV also showed increased expression of
NK-cell inhibitory receptors that prevent lysis of
target cells, but these effects were reversed in virally
suppressed patients on cART.85,86
No studies on NK
T-cell antibacterial defence in HIV-infected hosts were
identified, but selective NK T-cell depletion might
hinder the proinflammatory response to bacterial
pathogens.65
Various mechanisms contribute to expansion of an
anergic NK cell population that is less capable of
responding to pathogens in individuals with HIV. This
expansion might impair direct NK-cell mediated
defence against bacteria, and compromise crosstalk
with other immune cells.
Neutrophils
In patients with HIV infection, neutrophils might not
be able to function effectively because of several
mechanisms (appendix). First, neutropenia is common
in patients with HIV.87
Possible mechanisms include
increased apoptosis due to the presence of auto-
antibodies,88
activation in the absence of secondary
infection,89
reduced production attributed to decreased
levels of CSF3,90
and treatment-induced neutropenia.49
Additionally, defects in neutrophil function have been
described in patients with HIV, including reduced
chemotaxis,91,92
impaired transepithelial migration93
(possibly explained by reduced expression of
interleukin 8 receptors on neutrophils of these
patients92
), and impaired phagocytosis.40,94–96
In one
study96
phagocytosis of S aureus was only impaired
when neutrophils were incubated with serum of
patients with HIV, indicating reduced opsonisation
with antibodies as the mechanism underlying defective
phagocytosis. However, in other investigations,94,95
decreased phagocytosis was observed after previous
opsonisation in healthy serum, suggesting an antibody-
independent mechanism. Although the aforementioned
studies used neutrophils from patients with HIV with
advanced disease, one study43
recorded an increased
capacity for neutrophils to phagocytose E coli and
S aureus from patients with early HIV infection.
Superoxide production, a mechanism of microbial
killing, was increased in unstimulated neutrophils
from patients with HIV,43
but the response to pathogens,
such as E coli, was reduced.40,89
Additionally, HIV causes
impaired C5a (a complement constituent with
proinflammatory properties) and interleukin-8-induced
degranulation,97
which is associated with reduced
expression of C5a and interleukin 8 receptors.92,97
Although HIV causes NET release from neutrophils, it
can also counteract this response by inducing
production of interleukin 10 by dendritic cells to inhibit
NET formation.98
Hence, HIV-induced inhibition of
NET formation in response to bacterial pathogens
100 www.thelancet.com/infection Vol 15 January 2015
Review
could impair effective host defence. Finally, ex-vivo
bacterial killing was reduced,96
especially in patients
with advanced disease.96
However, one study94
reported
normal killing of S aureus by neutrophils from patients
with AIDS.
HIV-related defects in neutrophil numbers, chemo-
taxis, phagocytosis, superoxide production, cytokine
release, bacterial killing, and NET formation have been
described, suggesting that HIV renders the host more
susceptible to bacterial infection, at least in part by
impairing neutrophil-mediated host defence.
Cytokines and DAMPs
Abundant evidence suggests a change in cytokine
profiles in patients with HIV, although very few studies
have investigated this change in the context of sepsis
(table 2). Two studies50,51
provided comparative data for
inflammatory parameters in patients with sepsis
admitted to the ICU with or without HIV co-infection.
A wide range of cytokines were measured, but no
significant differences were noted for most of them. In
one study, high concentrations of interleukin 10 were
reported in HIV-positive patients compared with HIV-
negative patients, which was associated with increased
mortality.50
However, the HIV-negative septic controls
differed substantially in age and site of infection, which
complicates interpretation of these results.50,51
Studies47,99
using plasma from patients with HIV but
without sepsis reported increased concentrations of
several cytokines implicated in sepsis pathogenesis:
interleukin 6, interleukin 10, TNFα, interleukin 1A,
interleukin 1β, interleukin 8, CCL2, interleukin 1
receptor antagonist, and, in acute HIV infection,
interferon γ. Concentrations of HMGB1 were also
increased in patients with HIV.100
However, circulating
concentrations of the proinflammatory cytokines
interleukin 12, and CSF2, were decreased.49,48
Studies on
the effect of cART noted normalisation of most cytokine
perturbations.47,48
Raised concentrations of circulating inflammatory
markers might in fact be associated with the
development of infection; a correlation was identified
between raised concentrations of C-reactive protein in
patients with HIV and an increased risk of bacterial
pneumonia.101
A possible explanation for raised cytokine con-
centrations is a primed state of immune cells in patients
with HIV causing hyper-responsiveness to stimulation;
for example, by microbial products translocated from
the gut. Increased TNFα production in response to
lipopolysaccharide was observed in PBMCs and
monocytes isolated from patients with HIV.38,46
Decreased interleukin 12 concentrations might relate to
the type of stimulus and stage of disease, with patients
with AIDS showing the most profound deficiency.102
There is abundant evidence for increased circulating
concentrations of both proinflammatory and
anti-inflammatory cytokines in patients with HIV,
combined with ex-vivo evidence for a primed state of
immune cells in response to stimulation, which could
result in a more profound imbalance between these
mediators during sepsis. However, the small number of
studies available50,51
in patients with HIV and sepsis
reported few differences in cytokine concentrations
compared with HIV-negative patients with sepsis.
Lymphocytes
Overview
Antigen-presenting cells interact with lymphocytes to
initiate the adaptive immune response. In response to
the presentation of antigen, effector CD4 T cells secrete
cytokines, such as interferon γ, to enhance phagocytic
killing and interaction with B cells to initiate production
of antibodies.19
B cells are the cornerstone of immuno-
logical memory, and represent the main defence
mechanism against reinfection. B cells have also been
shown to have an important role in the enhancement of
innate immune responses during bacterial sepsis.103
Sepsis causes a substantial depletion of CD4, CD8, and
B lymphocytes because of increased apoptosis, which is
correlated with sepsis severity.17,104
Because T cells are
important in coordinating innate immunity, increased
apoptosis of T cells might hinder the proinflammatory
response.104
Remaining lymphocytes also show
significant reductions in cytokine release on
stimulation.105
T lymphocytes
During HIV infection, both T-cell numbers and
function become compromised (appendix). CD4 T-cell
depletion develops through various mechanisms,
including direct virus-induced killing, induction of
apoptosis, and an inability of the thymus to efficiently
compensate for the lost T cells.106
In addition to
depletion of CD4 T cells, HIV downregulates the CD4
receptor, which might reduce the immune functions of
surviving cells.107
Of particular relevance in the context
of sepsis, patients with HIV displayed increased
apoptosis of CD3 T cells after in-vitro challenge with
S pneumoniae.108
A particular subset of CD4 cells, Th17
cells, which predominate in the gastrointestinal tract
and are important in antibacterial defence, are
substantially depleted,109
and remaining Th17 cells have
an enahanced interleukin 10:TNFα ratio, suggestive of
an anti-inflammatory phenotype (table 2).67
Similarly,
increased interleukin 10 production was identified in
peripheral blood CD4 T cells.66
Mucosal-associated
invariant T cells (MAIT cells), a subset of innate-like,
tissue-infiltrating lymphocytes that produce
interleukin 17, interleukin 22, interferon γ, and TNFα,
were also shown to be severely depleted in blood of
patients with HIV, and remaining cells showed an
activated phenotype combined with functional
exhaustion.110,111
Although numbers of MAIT cells in the
www.thelancet.com/infection Vol 15 January 2015 101
Review
gut were relatively preserved in these studies,110,111
a
third study reported MAIT cell depletion in the colon of
patients with HIV.112
Since MAIT cells are known to be
activated by a wide range of bacteria and fungi,113
their
depletion might have an important role in increased
susceptibility of patients with HIV to bacterial sepsis.
Mucosal depletion of interleukin-17-producing cells,
like TH17 cells and MAIT cells, might be related to
increased risk of patients with HIV developing non-
typhoidal salmonella bacteraemia, because interleukin
17 is known to have an important role in host defence
against dissemination of enteric pathogens (figure 2).114
Additionally, interleukin-17-producing CD4 T cells were
shown to be essential for recruitment of monocytes and
macrophages to allow for effective pneumococcal
clearance from the nasopharynx,115
suggesting that CD4
cell depletion has a role in the vulnerability of patients
with HIV to the invasive pneumococcal infections
(figure 1).
Although CD4 T-cell counts decrease, CD8 T-cell
numbers expand during HIV infection.116
Nonetheless,
both T-cell compartments display a phenotype of
immune activation and exhaustion, as suggested by
heightened expression of the activation markers HLA-
DR and CD38, and of the inhibitory receptors PDCD1
and CTLA4 in untreated patients with HIV.117,118
Furthermore, the CD8 T-cell compartment has been
shown to arrest at a late differentiated phenotype,119
and
is unable to mount an effective cytolytic response.120,121
Both reduction in CD4 T-cell numbers, and functional
impairments of CD4 T cells and CD8 T cells might
contribute to increased susceptibility of patients with
HIV to develop invasive bacterial infections and sepsis.
Most of these defects can be restored by cART,106
but
impaired restoration of the CD4 T-cell proliferation
response has been described in patients with HIV with
lower nadir CD4 T-cell counts before therapy,122
thus
providing one possible explanation for higher rates of
bacteraemia in cART-treated patients compared with
HIV-negative people.
B-lymphocytes
In patients with HIV, several defects in B-cell function
have been detected, which could have a role in increased
susceptibility to infections, especially in patients with
non-typhoidal salmonella and S pneumoniae (appendix).
Functional disturbances include hypergammaglo-
bulinaemia, polyclonal activation and exhaustion,
impaired class-switch recombination, dysfunctional
interaction between T cells and B cells, blunted
proliferation response to both T-cell-dependent and
T-cell-independent antigens, poor immune responses
against vaccination antigens, short duration of antibody
response induced by pneumococcal vaccination, and
increased apoptosis.123–127
Additionally, patients with HIV show changes in the
distribution of B-cell subsets. Resting memory B cells
are severely depleted, whereas transitional B cells are
expanded.123
An aberrant B-cell population,
CD21low
CD27−
B cells, normally present in very low
numbers in peripheral blood, was reported to rise in
the blood of viraemic patients with HIV proportional to
viral load,128
which showed features of immune
activation and cellular exhaustion.129
Although changes
in most B-cell subtypes are restored by cART, resting
memory B cells usually remain depleted128
because they
can only be preserved when cART is started early in
infection.130
Additionally, IgG concentrations were
shown to remain raised despite long-term cART in 45%
of patients, indicating continuous B-cell activation.131
B-cell dysfunction probably has an important role in
the increased risk of patients with HIV developing
invasive infections with S pneumoniae (figure 1) and
non-typhoidal salmonella (figure 2). Patients with HIV
have reduced numbers of pneumococcal antigen-
specific IgG antibody-secreting cells and lower
concentrations of pneumococcal antibodies.124,132
Additionally, IgA in the epithelial lining fluid of the
lung showed impaired antipneumococcal activity.133
Although some studies reported normal,134
or even
Th17
↓Interleukin 17
Intestinal lumen
Lamina
propria
Bloodstream
Interleukin 17
NTS in the gut
of a host without
HIV infection
NTS in the
gut of an
HIV-infected
host
Th17 depletion
and reduced
interleukin 17
compromise
gut barrier
function
Inhibitory
antibodies bind
to LPS, which
prevents
bacterial killing
Binding of
IgG to outer
membrane
proteins
enables
bacterial lysis
and killing
C1q
LPS
MAC
Figure2: HIV-induced changes in host defence against non-typhoidal salmonella (NTS) infection fromthe gut
Normally, interleukin-17-producing Th17 cells effectively prevent invasion of NTS from the gut (left side), but
HIV infection results in Th17 cell depletion and reduced interleukin 17 concentrations, thereby compromising
the gut barrier function against NTS (right side). If NTS bacteria manage to reach the bloodstream in a host
without HIV infection, they are effectively opsonised by IgG antibodies that bind to the outer membrane
protein of NTS and enable complement (C1q) binding, followed by formation of a MAC on the bacterial surface,
resulting in lysis and killing of the bacteria (left side). By contrast, in the patient with HIV, NTS bacteria are
opsonised by antibodies that have an inhibitory effect on bacterial killing. They do not bind to the outer
membrane protein of the NTS, but to its LPS, which does not enable lysis and killing, and allows for survival of
NTS in the bloodstream (right side). NTS=non-typhoid salmonella. MAC=membrane attack complex.
LPS=lipopolysaccharide.
102 www.thelancet.com/infection Vol 15 January 2015
Review
increased concentrations of IgG anti-pneumococcal-
specific antibodies in patients with HIV,135
the opsonic
capacity of these antibodies was reduced.135
In the case
of NTS, IgG hypersecretion might be implicated in the
increased risk of infection in patients with HIV; serum
from African patients with HIV contained high
concentrations of IgG antibodies against non-typhoidal
salmonella that prevented destruction of Salmonella
spp.136
These antibodies had non-neutralising activity
against non-typhoidal salmonella lipopolysaccharide
and exerted a dose-dependent inhibitory effect on the
killing capacity of healthy serum, suggesting an
important role for anti-salmonella LPS IgG in the
prevention of clearance of non-typhoidal salmonella in
patients with HIV (figure 2).137
Perturbations in humoral immunity probably have a
substantial role in the increased susceptibility of
patients with HIV to bacterial infections, with
simultaneous depletion of the memory B-cell com-
partment, and inadequate activation, IgG production,
and B-cell exhaustion, which are only partly restored by
cART.
The coagulation system
Patients with sepsis invariably show a disturbed balance
between coagulation and anticoagulation.16,19
In severe
cases, this disturbance results in disseminated intra-
vascular coagulation, while at the same time, a
consumptive coagulopathy develops, causing increased
risk of bleeding. disseminated intravascular coagulation
is triggered by a fulminant host response to infection,
causing aberrant expression of tissue factor, impairment
of physiological anticoagulant pathways due to
endothelial dysfunction, and the suppression of
fibrinolysis due to overproduction of SERPINE1 by
endothelial cells.16
Several abnormalities in the coagulation system and
endothelial cell function of patients with HIV have
been identified (figure 3). In general, HIV-induced
alterations in haemostasis are remarkably similar to
those described in sepsis. First, patients with HIV
display a procoagulant state, as shown by their
increased risk of thromboembolic events.137
Coagulation
activation in these patients is further suggested by
increased concentrations of D-dimer and fibrinogen,138,139
increased platelet activation,140
and increased expression
of tissue factor on platelets and monocytes.141,142
Anticoagulation is hindered by reduced concentrations
of protein C, and protein S,138,143
and fibrinolysis might
be impaired as a consequence of increased SERPINE
concentrations in patients with HIV.139,143
Last, ample
evidence shows endothelial activation and dysfunction
in patients with HIV. Raised concentrations of VWF,
soluble ICAM1, and soluble VCAM1 have been reported
in several studies.138,143,144
Additionally, endothelial
function, assessed by measuring flow-mediated dilation
of the brachial artery, was impaired in patients with
HIV.145
Autopsy studies of patients with HIV, with or
without AIDS-related complications, also reported
endothelial injury.146,147
Finally, a study on angiogenic
factors in severe bacterial infection in Malawian
children noted that HIV-positive cases had significantly
higher concentrations of ANGPT2 compared with HIV-
negative children with similar illness.148
High con-
centrations of ANGPT2, an angiogenic peptide that
increases endothelial activation and vascular
permeability, are associated with disseminated intra-
vascular coagulation and mortality in sepsis.148
Studies
on the effect of cART are inconsistent, with some
recording complete normalisation144
and others
reporting partial improvement,136,149
as well as studies
describing a detrimental effect of cART, especially
regimens containing protease inhibitors.137,150
At present, no studies on the effect of HIV infection
on the procoagulant response to sepsis exist. In view of
the disturbed haemostatic balance in patients with HIV,
with alterations that strongly resemble those detected
in sepsis, it is conceivable that HIV causes an even
greater disruption of procoagulant and anticoagulant
mechanisms during sepsis.
The complement system
The complement system is an important part of the
first-line host defence against bacteria, by opsonisation,
lysis of pathogens and infected cells, and production of
chemoattractants. Clinical and experimental sepsis are
↑ANGPT2
↑sICAM1
↑sVCAM1 ↑VWF
↑Tissue factor
Coagulation cascade
↑Fibrinogen Fibrin
Thrombus formation↑D-dimer
Activated protein C
↓Protein C
↓Protein S
↑SERPINE
↓Fibrinolysis
Increased coagulation Impaired anticoagulation and fibrinolysis
Endothelial activation and damage
Figure3:HIV-inducedchangesinthecoagulationsystemandendotheliumresemblesepsis-inducedcoagulopathy
Expression of tissue factor on circulating monocytes is increased in patients with HIV, which triggers the
extrinsic pathway of the coagulation cascade, eventually resulting in the transformation of fibrinogen to fibrin.
Increased concentrations of fibrinogen and D-dimer, a fibrin degradation product, are seen in patients with HIV,
providing further evidence for coagulation activation. The coagulation cascade is normally balanced by the
anticoagulant system, which includes activated protein C. However, in patients with HIV protein C, and protein
S, which is needed to activate protein C, are depleted. Additionally, fibrinolysis is inhibited by increased
concentrations of SERPINE, further facilitating thrombus formation in patient with HIV. The endothelium of
patients with HIV becomes activated and damaged. Secretion of sICAM1, sVCAM1, and VWF are increased, and
the endothelium shows an irregular pattern with multinucleated endothelial cells and increased monocyte
adhesion. Higher concentrations of ANGPT2 are also seen in patients with HIV,and are known to cause
endothelial cell death and disrupt the endothelial monolayer. Notably, all these changes are very similar to those
described in patients with sepsis. sICAM-1=soluble ICAM1. sVCAM1=soluble VCAM1.
www.thelancet.com/infection Vol 15 January 2015 103
Review
associated with activation of the complement system,
as shown by increased plasma concentrations of com-
plement constituents C3a, C4a, and C5a.21
Although
complement is essential in the host defence against
bacteria, increased circulating C3a and C5a are likely to
contribute to sepsis-induced tissue damage, multiorgan
failure, and septic shock, because of their strong
proinflammatory functions.151
C5a is important for the
outcome of experimental sepsis, as reported by studies
in which treatment with an anti-C5a antibody improved
haemodynamic parameters, attenuated coagulopathy,
and improved organ function.21
Although the complement system is generally known
for its activity against bacterial pathogens, many reports
describe HIV-induced activation of the complement
system through the the classic pathway via interaction
of gp41 with C1q;152
interaction of HIV with mannose-
binding lectin, the triggering molecule of the lectin
pathway;152
and activation of the alternative pathway
through C3 binding of HIV-infected monocytes and
lymphocytes.153
After seroconversion, HIV-specific
antibodies further enhance complement activation via
the classic pathway,152
and raised concentrations of
circulating C5a have been recorded in patients with
HIV.91
However, HIV can escape complement-induced
lysis by acquiring regulators of complement activation,
which allow the virus to use the complement system for
transport to the lymphoid system and infection of
susceptible cells.152
Increased complement activation might contribute to
the generalised immune activation observed in HIV
and, hypothetically, have a detrimental role during
sepsis pathogenesis by disturbing the balance between
proinflammatory and anti-inflammatory mechanisms.
However, at present, studies investigating the effect of
HIV infection on complement activation in sepsis have
not been reported.
Future perspectives
The present treatment of sepsis is based on antibiotics
and supportive care.154
In past decades, many clinical
trials have been done to investigate inhibition of the
abundant inflammatory response generally held
responsible for sepsis mortality.19,21
In more recent
years, however, attention has shifted to findings that
patients with sepsis invariably show evidence for
immune suppression, which has been implicated as an
important cause of secondary infections and late
mortality.17
Preclinical studies have suggested that
immune stimulatory therapy, which aims to overcome
sepsis-induced immune suppression, could improve
sepsis outcome. Interventions assessed in this context
include immunomodulatory cytokines, such as
interleukin 7, interleukin 15, and CSF2, as well as
antibodies targeting co-inhibitory molecules, like
PDCD1, CD274, and CTLA417,155
(table 3).156,157
Notably,
similar strategies have been suggested to remedy
Relevance in sepsis Relevance in HIV
Interleukin 7 Prevented apoptosis-inducedT-cell
depletion, and reversed sepsis-induced
depressionofT-cell cytokines in a murine
sepsis model
Improved lymphocyte function after ex-vivo
treatmentof cells from septic patients
Increase in functional naive and central
memoryCD4 andCD8T cells in clinicaltrials
Interleukin 15 Induces proliferation and activationof
T cells, natural killer cells and natural killer
T cells
Decreased apoptosis and improved survival
in a murine sepsis model
Theoretically,the same beneficial effects as in
sepsis might beobserved; however,delayed
viral suppression and failureto reconstitute
CD4T cells in SIV-infected macaquestreated
with interleukin 15discouraged further
research into interleukin 15 as an
immunomodulatorydrug
CSF2 Intargeted patient groups (thosewith low
HLA-DR expression inone study, and low
ex-vivotumour necrosis factor α producing
capacity)CSF2 increasedthe functionof
innate immune cells and improved clinical
outcomes
CSF2duringART interruptions blunted viral
rebound and preventedthedecreaseofCD4
cell counts156
PDCD1/CD274 Blockageof PDCD1or its ligand improved
survival in murine sepsis studies
Blockageof PDCD1 causedCD8T-cell and
B-cell activation, reduced viral load, and
improved survival in an SIV-macaque model.
PDCD1 blockade reduced hyperimmune
activation and microbialtranslocation in a
SIV-macaque model
CD274 blockade causedCD4T-cell restoration
in an ex-vivo studyusing PBMCs from patients
with HIV157
CTLA4 Anti-CTLA4treatment improved sepsis-
induced lymphocyte apoptosis and survival
from secondary fungal infections in a
murine sepsis model
Anti-CTLA4 treatment caused an increase in
CD4 and CD8 T cell responses and reduced
viral RNA in a SIV-macaque model
However, a second study with a similar
design reported no expansion of SIV specific
T cells and increased viral replication at
mucosal sites
Information in this table was derived from Hotchkiss et al and Hutchins et al17,155
for sepsis, andVanham andVan Gulck
for HIV,158
unless otherwise indicated in the table. ART=antiretroviral therapy. PBMC=peripheral blood mononuclear
cell. SIV=simian immunodeficiency virus.
Table 3: Parallels between the use of immunomodulatory agents in sepsis and HIV
Search strategy and selection criteria
We searched PubMed using the following search string:
(“HIV” [MeSH Terms] or “acquired immunodeficiency
syndrome” [MeSH Terms]) and (“immunity” [MeSH Terms]
or “monocytes” [MeSH Terms] or “macrophages” [MeSH
Terms] or “dendritic cell” [MeSH Terms] or “neutrophils”
[MeSH Terms] or “pattern recognition receptors” [MeSH
Terms] or “lymphocytes” [MeSH Terms] or “cytokines”
[MeSH Terms] or “complement system proteins” [MeSH
Terms] or “blood coagulation” [MeSH Terms] or
“disseminated intravascular coagulation” [MeSH Terms])
and (“bacteria” [MeSH Terms] or “sepsis” [MesH Terms] or
“bacteremia” [Mesh Terms)”. We also combined the MeSH
Terms for “salmonella” and “S pneumoniae” with the MeSH
Terms for “HIV” or “AIDS”. The search was limited to
English-language articles published between Jan 1, 1990
and June 1, 2014. We reviewed relevant articles identified in
this search, papers cited in these studies, and articles from
the authors’ personal files.
104 www.thelancet.com/infection Vol 15 January 2015
Review
HIV-induced immune suppression (table 3).158
Pro-
inflammatory and procoagulant pathways targeted by
novel sepsis therapies under clinical evaluation, such as
anti-HMGB1 strategies and soluble thrombo-
modulin,159,160
could be of interest for patients with HIV
and sepsis because HIV by itself already adversely
affects these mechanisms. Many pathogenic mecha-
nisms between sepsis and HIV overlap, so it is
conceivable that patients with HIV presenting with
sepsis form a group that will particularly benefit from
new drugs targeting septic immunosuppression. In
this light, the systematic exclusion of patients with HIV
from sepsis trials might need to be reconsidered.
Conclusion
In the era of effective cART, bacterial sepsis has evolved
as a major cause of mortality in patients with HIV. HIV
infection affects many components of the immune
response, which on the one hand renders the HIV-
infected host more susceptible to invasive infection, and
on the other hand might further exacerbate the
deregulated host response to sepsis. Specific research on
sepsis in patients with HIV is scarce and future studies
are needed to gain more insight into the particularities of
sepsis pathogenesis in these patients. Although cART
greatly improves patients’ immune function, as shown
by the strong reduction in opportunistic infections,
patients continue to have an increased risk of developing
invasive bacterial infection, suggesting the current and
future relevance of this under-researched area.
Declaration of interests
We declare no competing interests.
Contributors
MAH conceived the idea, searched the scientific literature, interpreted
the data, wrote the manuscript and created the figures. MPG and
TvdP. interpreted the data and wrote the manuscript.
References
1 Kim JH, Psevdos G Jr, Gonzalez E, Singh S, Kilayko MC, Sharp V.
All-cause mortality in hospitalized HIV-infected patients at an
acute tertiary care hospital with a comprehensive outpatient HIV
care program in New York City in the era of highly active
antiretroviral therapy (HAART). Infection 2013; 41: 545–51.
2 Gilks CF, Ojoo SA, Ojoo JC, et al. Invasive pneumococcal disease
in a cohort of predominantly HIV-1 infected female sex-workers in
Nairobi, Kenya. Lancet 1996; 347: 718–23.
3 Huson MA, Stolp SM, van der Poll T, Grobusch MP. Community-
acquired bacterial bloodstream infections in HIV-infected
patients: a systematic review. Clin Infect Dis 2014; 58: 79–92.
4 Jordano Q, Falcó V, Almirante B, et al. Invasive pneumococcal
disease in patients infected with HIV: still a threat in the era of
highly active antiretroviral therapy. Clin Infect Dis 2004; 38: 1623–28.
5 Archibald LK, den Dulk MO, Pallangyo KJ, Reller LB. Fatal
Mycobacterium tuberculosis bloodstream infections in febrile
hospitalized adults in Dar es Salaam, Tanzania. Clin Infect Dis
1998; 26: 290–96.
6 Archibald LK, McDonald LC, Nwanyanwu O, et al. A hospital-
based prevalence survey of bloodstream infections in febrile
patients in Malawi: implications for diagnosis and therapy.
J Infect Dis 2000; 181: 1414–20.
7 Arthur G, Nduba VN, Kariuki SM, Kimari J, Bhatt SM, Gilks CF.
Trends in bloodstream infections among human immunodeficiency
virus-infected adults admitted to a hospital in Nairobi, Kenya,
during the last decade. Clin Infect Dis 2001; 33: 248–56.
8 Bell M, Archibald LK, Nwanyanwu O, et al. Seasonal variation in
the etiology of bloodstream infections in a febrile inpatient
population in a developing country. Int J Infect Dis 2001; 5: 63–69.
9 Crump JA, Ramadhani HO, Morrissey AB, et al. Invasive bacterial
and fungal infections among hospitalized HIV-infected and HIV-
uninfected adults and adolescents in northern Tanzania.
Clin Infect Dis 2011; 52: 341–48.
10 Grant AD, Djomand G, Smets P, et al. Profound
immunosuppression across the spectrum of opportunistic disease
among hospitalized HIV-infected adults in Abidjan, Côte d’Ivoire.
AIDS 1997; 11: 1357–64.
11 Mayanja BN, Todd J, Hughes P, et al. Septicaemia in a population-
based HIV clinical cohort in rural Uganda, 1996–2007: incidence,
aetiology, antimicrobial drug resistance and impact of
antiretroviral therapy. Trop Med Int Health 2010; 15: 697–705.
12 Meremo A, Mshana SE, Kidenya BR, Kabangila R, Peck R,
Kataraihya JB. High prevalence of Non-typhoid salmonella
bacteraemia among febrile HIV adult patients admitted at a
tertiary Hospital, North-Western Tanzania. Int Arch Med
2012; 5: 28.
13 Nadjm B, Mtove G, Amos B, et al. Severe febrile illness in adult
hospital admissions in Tanzania: a prospective study in an area of
high malaria transmission. Trans R Soc Trop Med Hyg 2012;
106: 688–95.
14 Peters RP, Zijlstra EE, Schijffelen MJ, et al. A prospective study of
bloodstream infections as cause of fever in Malawi: clinical
predictors and implications for management. Trop Med Int Health
2004; 9: 928–34.
15 Ssali FN, Kamya MR, Wabwire-Mangen F, et al. A prospective
study of community-acquired bloodstream infections among
febrile adults admitted to Mulago Hospital in Kampala, Uganda.
J Acquir Immune Defic Syndr Hum Retrovirol 1998; 19: 484–89.
16 Angus DC, van der Poll T. Severe sepsis and septic shock.
N Engl J Med 2013; 369: 840–51.
17 Hotchkiss RS, Monneret G, Payen D. Immunosuppression in
sepsis: a novel understanding of the disorder and a new
therapeutic approach. Lancet Infect Dis 2013; 13: 260–68.
18 Appay V, Sauce D. Immune activation and inflammation in HIV-1
infection: causes and consequences. J Pathol 2008; 214: 231–41.
19 Stearns-Kurosawa DJ, Osuchowski MF, Valentine C, Kurosawa S,
Remick DG. The pathogenesis of sepsis. Annu Rev Pathol 2011;
6: 19–48.
20 Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate
immunity. Cell 2006; 124: 783–801.
21 van der Poll T, Opal SM. Host-pathogen interactions in sepsis.
Lancet Infect Dis 2008; 8: 32–43.
22 Mancuso G, Gambuzza M, Midiri A, et al. Bacterial recognition by
TLR7 in the lysosomes of conventional dendritic cells.
Nat Immunol 2009; 10: 587–94.
23 Cavassani KA, Ishii M, Wen H, et al. TLR3 is an endogenous
sensor of tissue necrosis during acute inflammatory events.
J Exp Med 2008; 205: 2609–21.
24 Murphy KM. Janeway’s immunobiology, 8th edn. New York:
Garland Science; 2012.
25 Chiche L, Forel JM, Thomas G, et al. The role of natural killer
cells in sepsis. J Biomed Biotechnol 2011: 986491.
26 Souza-Fonseca-Guimaraes F, Adib-Conquy M, Cavaillon JM.
Natural killer (NK) cells in antibacterial innate immunity: angels
or devils? Mol Med 2012; 18: 270–85.
27 Leung B, Harris HW. NKT cells: the culprits of sepsis? J Surg Res
2011; 167: 87–95.
28 Nathan C. Neutrophils and immunity: challenges and
opportunities. Nat Rev Immunol 2006; 6: 173–82.
29 de Jong HK, van der Poll T, Wiersinga WJ. The systemic pro-
inflammatory response in sepsis. J Innate Immun 2010; 2: 422–30.
30 Yang H, Antoine DJ, Andersson U, Tracey KJ. The many faces of
HMGB1: molecular structure-functional activity in inflammation,
apoptosis, and chemotaxis. J Leukoc Biol 2013; 93: 865–73.
31 Epple HJ, Zeitz M. HIV infection and the intestinal mucosal
barrier. Ann N Y Acad Sci 2012; 1258: 19–24.
32 Brenchley JM, Price DA, Schacker TW, et al. Microbial
translocation is a cause of systemic immune activation in chronic
HIV infection. Nat Med 2006; 12: 1365–71.
www.thelancet.com/infection Vol 15 January 2015 105
Review
33 Sanders CM, Cruse JM, Lewis RE. Toll-like receptors, cytokines
and HIV-1. Exp Mol Pathol 2008; 84: 31–36.
34 Scagnolari C, Selvaggi C, Chiavuzzo L, et al. Expression levels of
TLRs involved in viral recognition in PBMCs from HIV-1-infected
patients failing antiretroviral therapy. Intervirology 2009;
52: 107–14.
35 Hernández JC, Stevenson M, Latz E, Urcuqui-Inchima S. HIV
type 1 infection up-regulates TLR2 and TLR4 expression and
function in vivo and in vitro. AIDS Res Hum Retroviruses 2012;
28: 1313–28.
36 Heggelund L, Müller F, Lien E, et al. Increased expression of toll-
like receptor 2 on monocytes in HIV infection: possible roles in
inflammation and viral replication. Clin Infect Dis 2004; 39:
264–69.
37 Gordon SB, Jagoe RT, Jarman ER, et al. The alveolar
microenvironment of patients infected with human
immunodeficiency virus does not modify alveolar macrophage
interactions with Streptococcus pneumoniae.
Clin Vaccine Immunol 2013; 20: 882–91.
38 Lester RT, Yao XD, Ball TB, et al. Toll-like receptor expression and
responsiveness are increased in viraemic HIV-1 infection. AIDS
2008; 22: 685–94.
39 Schuitemaker H, Kootstra NA, Fouchier RA, Hooibrink B,
Miedema F. Productive HIV-1 infection of macrophages restricted
to the cell fraction with proliferative capacity. EMBO J 1994;
13: 5929–36.
40 Michailidis C, Giannopoulos G, Vigklis V, Armenis K, Tsakris A,
Gargalianos P. Impaired phagocytosis among patients infected by
the human immunodeficiency virus: implication for a role of
highly active anti-retroviral therapy. Clin Exp Immunol 2012;
167: 499–504.
41 Trial J, Birdsall HH, Hallum JA, et al. Phenotypic and functional
changes in peripheral blood monocytes during progression of
human immunodeficiency virus infection. Effects of soluble
immune complexes, cytokines, subcellular particulates from
apoptotic cells, and HIV-1-encoded proteins on monocytes
phagocytic function, oxidative burst, transendothelial migration,
and cell surface phenotype. J Clin Invest 1995; 95: 1690–701.
42 Nottet HS, de Graaf L, de Vos NM, et al. Phagocytic function of
monocyte-derived macrophages is not affected by human
immunodeficiency virus type 1 infection. J Infect Dis 1993;
168: 84–91.
43 Bandres JC, Trial J, Musher DM, Rossen RD. Increased
phagocytosis and generation of reactive oxygen products by
neutrophils and monocytes of men with stage 1 human
immunodeficiency virus infection. J Infect Dis 1993; 168: 75–83.
44 Brown JN, Kohler JJ, Coberley CR, Sleasman JW, Goodenow MM.
HIV-1 activates macrophages independent of Toll-like receptors.
PLoS One 2008; 3: e3664.
45 Gekonge B, Giri MS, Kossenkov AV, et al. Constitutive gene
expression in monocytes from chronic HIV-1 infection overlaps
with acute Toll-like receptor induced monocyte activation profiles.
PLoS One 2012; 7: e41153.
46 Dutertre CA, Amraoui S, DeRosa A, et al. Pivotal role of M-DC8+
monocytes from viremic HIV-infected patients in TNFα
overproduction in response to microbial products. Blood 2012;
120: 2259–68.
47 Haissman JM, Vestergaard LS, Sembuche S, et al. Plasma
cytokine levels in Tanzanian HIV-1-infected adults and the effect
of antiretroviral treatment. J Acquir Immune Defic Syndr 2009;
52: 493–97.
48 Keating SM, Golub ET, Nowicki M, et al, and the Women’s
Interagency HIV Study. The effect of HIV infection and HAART
on inflammatory biomarkers in a population-based cohort of
women. AIDS 2011; 25: 1823–32.
49 Kuritzkes DR. Neutropenia, neutrophil dysfunction, and bacterial
infection in patients with human immunodeficiency virus
disease: the role of granulocyte colony-stimulating factor.
Clin Infect Dis 2000; 30: 256–60.
50 Silva JM Jr, dos Santos SS. Sepsis in AIDS patients: clinical,
etiological and inflammatory characteristics. J Int AIDS Soc 2013;
16: 17344.
51 Amancio RT, Japiassu AM, Gomes RN, et al. The innate immune
response in HIV/AIDS septic shock patients: a comparative study.
PLoS One 2013; 8: e68730.
52 Lester RT, Yao XD, Ball TB, et al. HIV-1 RNA dysregulates the
natural TLR response to subclinical endotoxemia in Kenyan
female sex-workers. PLoS One 2009; 4: e5644.
53 Tachado SD, Zhang J, Zhu J, Patel N, Koziel H. HIV impairs
TNF-alpha release in response to Toll-like receptor 4 stimulation
in human macrophages in vitro. Am J Respir Cell Mol Biol 2005;
33: 610–21.
54 Nicol MQ, Mathys JM, Pereira A, Ollington K, Ieong MH,
Skolnik PR. Human immunodeficiency virus infection alters
tumor necrosis factor alpha production via Toll-like receptor-
dependent pathways in alveolar macrophages and U1 cells. J Virol
2008; 82: 7790–98.
55 Gordon SB, Jarman ER, Kanyanda S, et al. Reduced interleukin-8
response to StreptococcuS pneumoniae by alveolar macrophages
from adults with HIV/AIDS. AIDS 2005; 19: 1197–200.
56 Denis M, Ghadirian E. Dysregulation of interleukin 8, interleukin
10, and interleukin 12 release by alveolar macrophages from HIV
type 1-infected subjects. AIDS Res Hum Retroviruses 1994;
10: 1619–27.
57 Smed-Sörensen A, Loré K, Walther-Jallow L, Andersson J,
Spetz AL. HIV-1-infected dendritic cells up-regulate cell surface
markers but fail to produce IL-12 p70 in response to CD40 ligand
stimulation. Blood 2004; 104: 2810–17.
58 Frleta D, Ochoa CE, Kramer HB, et al. HIV-1 infection-induced
apoptotic microparticles inhibit human DCs via CD44.
J Clin Invest 2012; 122: 4685–97.
59 Benlahrech A, Gotch F, Kelleher P, Patterson S. Loss of NK
stimulatory capacity by plasmacytoid and monocyte-derived DC
but not myeloid DC in HIV-1 infected patients. PLoS One 2011;
6: e17525.
60 Donaghy H, Gazzard B, Gotch F, Patterson S. Dysfunction and
infection of freshly isolated blood myeloid and plasmacytoid
dendritic cells in patients infected with HIV-1. Blood 2003;
101: 4505–11.
61 Feldman S, Stein D, Amrute S, et al. Decreased interferon-alpha
production in HIV-infected patients correlates with numerical and
functional deficiencies in circulating type 2 dendritic cell
precursors. Clin Immunol 2001; 101: 201–10.
62 Chougnet C, Cohen SS, Kawamura T, et al. Normal immune
function of monocyte-derived dendritic cells from HIV-infected
individuals: implications for immunotherapy. J Immunol 1999;
163: 1666–73.
63 Sabado RL, O’Brien M, Subedi A, et al. Evidence of dysregulation
of dendritic cells in primary HIV infection. Blood 2010;
116: 3839–52.
64 Dillon SM, Lee EJ, Bramante JM, Barker E, Wilson CC. The
natural killer cell interferon-gamma response to bacteria is
diminished in untreated HIV-1 infection and defects persist
despite viral suppression. J Acquir Immune Defic Syndr 2014;
65: 259–67.
65 Vasan S, Tsuji M. A double-edged sword: the role of NKT cells in
malaria and HIV infection and immunity. Semin Immunol 2010;
22: 87–96.
66 Ostrowski MA, Gu JX, Kovacs C, Freedman J, Luscher MA,
MacDonald KS. Quantitative and qualitative assessment of
human immunodeficiency virus type 1 (HIV-1)-specific CD4+
T cell immunity to gag in HIV-1-infected individuals with
differential disease progression: reciprocal interferon-gamma and
interleukin-10 responses. J Infect Dis 2001; 184: 1268–78.
67 Kim CJ, McKinnon LR, Kovacs C, et al. Mucosal Th17 cell
function is altered during HIV infection and is an independent
predictor of systemic immune activation. J Immunol 2013;
191: 2164–73.
68 Jambo KC, Banda DH, Kankwatira AM, et al. Small alveolar
macrophages are infected preferentially by HIV and exhibit
impaired phagocytic function. Mucosal Immunol 2014;
7: 1116–26.
69 van der Poll T, Keogh CV, Buurman WA, Lowry SF. Passive
immunization against tumor necrosis factor-alpha impairs host
defense during pneumococcal pneumonia in mice.
Am J Respir Crit Care Med 1997; 155: 603–08.
70 Olliver M, Hiew J, Mellroth P, Henriques-Normark B, Bergman P.
Human monocytes promote Th1 and Th17 responses to
Streptococcus pneumoniae. Infect Immun 2011; 79: 4210–17.
106 www.thelancet.com/infection Vol 15 January 2015
Review
71 Dockrell DH, Marriott HM, Prince LR, et al. Alveolar
macrophage apoptosis contributes to pneumococcal clearance in
a resolving model of pulmonary infection. J Immunol 2003;
171: 5380–88.
72 Patel NR, Zhu J, Tachado SD, et al. HIV impairs TNF-alpha
mediated macrophage apoptotic response to Mycobacterium
tuberculosis. J Immunol 2007; 179: 6973–80.
73 Dillon SM, Robertson KB, Pan SC, et al. Plasmacytoid and
myeloid dendritic cells with a partial activation phenotype
accumulate in lymphoid tissue during asymptomatic chronic
HIV-1 infection. J Acquir Immune Defic Syndr 2008; 48: 1–12.
74 Donaghy H, Pozniak A, Gazzard B, et al. Loss of blood CD11c(+)
myeloid and CD11c(-) plasmacytoid dendritic cells in patients with
HIV-1 infection correlates with HIV-1 RNA virus load. Blood 2001;
98: 2574–76.
75 Loré K, Sönnerborg A, Broström C, et al. Accumulation of
DC-SIGN+CD40+ dendritic cells with reduced CD80 and CD86
expression in lymphoid tissue during acute HIV-1 infection.
AIDS 2002; 16: 683–92.
76 Dillon SM, Friedlander LJ, Rogers LM, et al. Blood myeloid
dendritic cells from HIV-1-infected individuals display a
proapoptotic profile characterized by decreased Bcl-2 levels and by
caspase-3+ frequencies that are associated with levels of plasma
viremia and T cell activation in an exploratory study. J Virol 2011;
85: 397–409.
77 Izmailova E, Bertley FM, Huang Q, et al. HIV-1 Tat reprograms
immature dendritic cells to express chemoattractants for activated
T cells and macrophages. Nat Med 2003; 9: 191–97.
78 Granelli-Piperno A, Golebiowska A, Trumpfheller C, Siegal FP,
Steinman RM. HIV-1-infected monocyte-derived dendritic cells do
not undergo maturation but can elicit IL-10 production and T cell
regulation. Proc Natl Acad Sci USA 2004; 101: 7669–74.
79 Yonkers NL, Rodriguez B, Asaad R, Lederman MM, Anthony DD.
Systemic immune activation in HIV infection is associated with
decreased MDC responsiveness to TLR ligand and inability to
activate naive CD4 T-cells. PLoS One 2011; 6: e23884.
80 Funke J, Dürr R, Dietrich U, Koch J. Natural killer cells in HIV-1
infection: a double-edged sword. AIDS Rev 2011; 13: 67–76.
81 Azzoni L, Papasavvas E, Chehimi J, et al. Sustained impairment
of IFN-gamma secretion in suppressed HIV-infected patients
despite mature NK cell recovery: evidence for a defective
reconstitution of innate immunity. J Immunol 2002;
168: 5764–70.
82 Jost S, Altfeld M. Evasion from NK cell-mediated immune
responses by HIV-1. Microbes Infect 2012; 14: 904–15.
83 Mavilio D, Lombardo G, Kinter A, et al. Characterization of the
defective interaction between a subset of natural killer cells and
dendritic cells in HIV-1 infection. J Exp Med 2006; 203: 2339–50.
84 Kottilil S, Chun TW, Moir S, et al. Innate immunity in human
immunodeficiency virus infection: effect of viremia on natural
killer cell function. J Infect Dis 2003; 187: 1038–45.
85 Kottilil S, Shin K, Planta M, et al. Expression of chemokine and
inhibitory receptors on natural killer cells: effect of immune
activation and HIV viremia. J Infect Dis 2004; 189: 1193–98.
86 Mavilio D, Benjamin J, Daucher M, et al. Natural killer cells in
HIV-1 infection: dichotomous effects of viremia on inhibitory and
activating receptors and their functional correlates.
Proc Natl Acad Sci USA 2003; 100: 15011–16.
87 Munyazesa E, Emile I, Mutimura E, et al. Assessment of
haematological parameters in HIV-infected and uninfected
Rwandan women: a cross-sectional study. BMJ Open 2012;
2: e001600.
88 Rubinstein DB, Farrington GK, O’Donnell C, Hartman KR,
Wright DG. Autoantibodies to leukocyte alphaMbeta2 integrin
glycoproteins in HIV infection. Clin Immunol 1999; 90: 352–59.
89 Elbim C, Prevot MH, Bouscarat F, et al. Polymorphonuclear
neutrophils from human immunodeficiency virus-infected
patients show enhanced activation, diminished fMLP-induced
L-selectin shedding, and an impaired oxidative burst after
cytokine priming. Blood 1994; 84: 2759–66.
90 Mauss S, Steinmetz HT, Willers R, et al. Induction of granulocyte
colony-stimulating factor by acute febrile infection but not by
neutropenia in HIV-seropositive individuals.
J Acquir Immune Defic Syndr Hum Retrovirol 1997; 14: 430–34.
91 Meddows-Taylor S, Pendle S, Tiemessen CT. Altered expression of
CD88 and associated impairment of complement 5a-induced
neutrophil responses in human immunodeficiency virus type
1-infected patients with and without pulmonary tuberculosis.
J Infect Dis 2001; 183: 662–65.
92 Meddows-Taylor S, Martin DJ, Tiemessen CT. Reduced expression
of interleukin-8 receptors A and B on polymorphonuclear
neutrophils from persons with human immunodeficiency virus
type 1 disease and pulmonary tuberculosis. J Infect Dis 1998;
177: 921–30.
93 Hofman P, Fischer F, Far DF, et al. Impairment of HIV
polymorphonuclear leukocyte transmigration across T84 cell
monolayers: an alternative mechanisms for increased intestinal
bacterial infections in AIDS? Eur Cytokine Netw 1999; 10: 373–82.
94 Pos O, Stevenhagen A, Meenhorst PL, Kroon FP, Van Furth R.
Impaired phagocytosis of StaphylococcuS aureus by granulocytes
and monocytes of AIDS patients. Clin Exp Immunol 1992;
88: 23–28.
95 Schaumann R, Krosing J, Shah PM. Phagocytosis of Escherichia
coli and StaphylococcuS aureus by neutrophils of human
immunodeficiency virus-infected patients. Eur J Med Res 1998;
3: 546–48.
96 Roilides E, Mertins S, Eddy J, Walsh TJ, Pizzo PA, Rubin M.
Impairment of neutrophil chemotactic and bactericidal function
in children infected with human immunodeficiency virus type 1
and partial reversal after in vitro exposure to granulocyte-
macrophage colony-stimulating factor. J Pediatr 1990; 117: 531–40.
97 Meddows-Taylor S, Kuhn L, Meyers TM, Sherman G,
Tiemessen CT. Defective neutrophil degranulation induced by
interleukin-8 and complement 5a and down-regulation of
associated receptors in children vertically infected with human
immunodeficiency virus type 1. Clin Diagn Lab Immunol 2001;
8: 21–30.
98 Saitoh T, Komano J, Saitoh Y, et al. Neutrophil extracellular traps
mediate a host defense response to human immunodeficiency
virus-1. Cell Host Microbe 2012; 12: 109–16.
99 Keating SM, Jacobs ES, Norris PJ. Soluble mediators of
inflammation in HIV and their implications for therapeutics and
vaccine development. Cytokine Growth Factor Rev 2012; 23:
193–206.
100 Nowak P, Barqasho B, Sönnerborg A. Elevated plasma levels of
high mobility group box protein 1 in patients with HIV-1
infection. AIDS 2007; 21: 869–71.
101 Bjerk SM, Baker JV, Emery S, et al, and the INSIGHT SMART
Study Group. Biomarkers and bacterial pneumonia risk in
patients with treated HIV infection: a case-control study. PLoS one
2013; 8: e56249.
102 Vanham G, Penne L, Devalck J, et al. Decreased CD40 ligand
induction in CD4 T cells and dysregulated IL-12 production
during HIV infection. Clin Exp Immunol 1999; 117: 335–42.
103 Kelly-Scumpia KM, Scumpia PO, Weinstein JS, et al. B cells
enhance early innate immune responses during bacterial sepsis.
J Exp Med 2011; 208: 1673–82.
104 Kasten KR, Tschöp J, Adediran SG, Hildeman DA, Caldwell CC.
T cells are potent early mediators of the host response to sepsis.
Shock 2010; 34: 327–36.
105 Boomer JS, To K, Chang KC, et al. Immunosuppression in
patients who die of sepsis and multiple organ failure. JAMA 2011;
306: 2594–605.
106 Gougeon ML. To kill or be killed: how HIV exhausts the immune
system. Cell Death Differ 2005; 12 (suppl 1): 845–54.
107 Piguet V, Schwartz O, Le Gall S, Trono D. The downregulation of
CD4 and MHC-I by primate lentiviruses: a paradigm for the
modulation of cell surface receptors. Immunol Rev 1999; 168:
51–63.
108 Daigneault M, De Silva TI, Bewley MA, et al. Monocytes regulate
the mechanism of T-cell death by inducing Fas-mediated apoptosis
during bacterial infection. PLoS Pathog 2012; 8: e1002814.
109 Klatt NR, Brenchley JM. Th17 cell dynamics in HIV infection.
Curr Opin HIV AIDS 2010; 5: 135–40.
110 Leeansyah E, Ganesh A, Quigley MF, et al. Activation, exhaustion,
and persistent decline of the antimicrobial MR1-restricted MAIT-
cell population in chronic HIV-1 infection. Blood 2013;
121: 1124–35.
www.thelancet.com/infection Vol 15 January 2015 107
Review
111 Cosgrove C, Ussher JE, Rauch A, et al. Early and nonreversible
decrease of CD161++ /MAIT cells in HIV infection. Blood 2013;
121: 951–61.
112 Greathead L, Metcalf R, Gazzard B, Gotch F, Steel A, Kelleher P.
CD8+/CD161++ mucosal-associated invariant T-cell levels in the
colon are restored on long-term antiretroviral therapy and
correlate with CD8+ T-cell immune activation. AIDS 2014;
28: 1690–92.
113 Gold MC, Lewinsohn DM. Co-dependents: MR1-restricted
MAIT cells and their antimicrobial function. Nat Rev Microbiol
2013; 11: 14–19.
114 Rubino SJ, Geddes K, Girardin SE. Innate IL-17 and IL-22
responses to enteric bacterial pathogens. Trends Immunol 2012;
33: 112–18.
115 Zhang Z, Clarke TB, Weiser JN. Cellular effectors mediating
Th17-dependent clearance of pneumococcal colonization in mice.
J Clin Invest 2009; 119: 1899–909.
116 Catalfamo M, Wilhelm C, Tcheung L, et al. CD4 and CD8 T cell
immune activation during chronic HIV infection: roles of
homeostasis, HIV, type I IFN, and IL-7. J Immunol 2011;
186: 2106–16.
117 Sachdeva M, Fischl MA, Pahwa R, Sachdeva N, Pahwa S.
Immune exhaustion occurs concomitantly with immune
activation and decrease in regulatory T cells in viremic
chronically HIV-1-infected patients. J Acquir Immune Defic Syndr
2010; 54: 447–54.
118 Larsson M, Shankar EM, Che KF, et al. Molecular signatures of
T-cell inhibition in HIV-1 infection. Retrovirology 2013; 10: 31.
119 Champagne P, Ogg GS, King AS, et al. Skewed maturation of
memory HIV-specific CD8 T lymphocytes. Nature 2001;
410: 106–11.
120 Appay V, Nixon DF, Donahoe SM, et al. HIV-specific CD8(+)
T cells produce antiviral cytokines but are impaired in cytolytic
function. J Exp Med 2000; 192: 63–75.
121 Andersson J, Behbahani H, Lieberman J, et al. Perforin is not
co-expressed with granzyme A within cytotoxic granules in CD8
T lymphocytes present in lymphoid tissue during chronic HIV
infection. AIDS 1999; 13: 1295–303.
122 Lange CG, Valdez H, Medvik K, Asaad R, Lederman MM. CD4+
T-lymphocyte nadir and the effect of highly active antiretroviral
therapy on phenotypic and functional immune restoration in
HIV-1 infection. Clin Immunol 2002; 102: 154–61.
123 Moir S, Fauci AS. Insights into B cells and HIV-specific B-cell
responses in HIV-infected individuals. Immunol Rev 2013;
254: 207–24.
124 Hart M, Steel A, Clark SA, et al. Loss of discrete memory B cell
subsets is associated with impaired immunization responses in
HIV-1 infection and may be a risk factor for invasive
pneumococcal disease. J Immunol 2007; 178: 8212–20.
125 Qiao X, He B, Chiu A, Knowles DM, Chadburn A, Cerutti A.
Human immunodeficiency virus 1 Nef suppresses CD40-
dependent immunoglobulin class switching in bystander B cells.
Nat Immunol 2006; 7: 302–10.
126 Nielsen H, Kvinesdal B, Benfield TL, Lundgren JD,
Konradsen HB. Rapid loss of specific antibodies after
pneumococcal vaccination in patients with human
immunodeficiency virus-1 infection. Scand J Infect Dis 1998; 30:
597–601.
127 Malaspina A, Moir S, Kottilil S, et al. Deleterious effect of HIV-1
plasma viremia on B cell costimulatory function. J Immunol 2003;
170: 5965–72.
128 Pensieroso S, Galli L, Nozza S, et al. B-cell subset alterations and
correlated factors in HIV-1 infection. AIDS 2013; 27: 1209–17.
129 Moir S, Ho J, Malaspina A, et al. Evidence for HIV-associated
B cell exhaustion in a dysfunctional memory B cell compartment
in HIV-infected viremic individuals. J Exp Med 2008; 205: 1797–805.
130 Moir S, Buckner CM, Ho J, et al. B cells in early and chronic HIV
infection: evidence for preservation of immune function
associated with early initiation of antiretroviral therapy. Blood
2010; 116: 5571–79.
131 Jacobson MA, Khayam-Bashi H, Martin JN, Black D, Ng V. Effect
of long-term highly active antiretroviral therapy in restoring HIV-
induced abnormal B-lymphocyte function.
J Acquir Immune Defic Syndr 2002; 31: 472–77.
132 Iwajomo OH, Finn A, Moons P, et al. Deteriorating
pneumococcal-specific B-cell memory in minimally symptomatic
African children with HIV infection. J Infect Dis 2011; 204: 534–43.
133 Moja P, Jalil A, Quesnel A, et al. Humoral immune response
within the lung in HIV-1 infection. Clin Exp Immunol 1997;
110: 341–48.
134 Gordon SB, Miller DE, Day RB, et al. Pulmonary immunoglobulin
responses to StreptococcuS pneumoniae are altered but not
reduced in human immunodeficiency virus-infected Malawian
adults. J Infect Dis 2003; 188: 666–70.
135 Takahashi H, Oishi K, Yoshimine H, et al. Decreased serum
opsonic activity against StreptococcuS pneumoniae in human
immunodeficiency virus-infected Ugandan adults. Clin Infect Dis
2003; 37: 1534–40.
136 MacLennan CA, Gilchrist JJ, Gordon MA, et al. Dysregulated
humoral immunity to nontyphoidal Salmonella in HIV-infected
African adults. Science 2010; 328: 508–12.
137 Matta F, Yaekoub AY, Stein PD. Human immunodeficiency virus
infection and risk of venous thromboembolism. Am J Med Sci
2008; 336: 402–06.
138 Jong E, Louw S, van Gorp EC, Meijers JC, ten Cate H, Jacobson BF.
The effect of initiating combined antiretroviral therapy on
endothelial cell activation and coagulation markers in South African
HIV-infected individuals. Thromb Haemost 2010; 104: 1228–34.
139 Koppel K, Bratt G, Schulman S, Bylund H, Sandström E.
Hypofibrinolytic state in HIV-1-infected patients treated with
protease inhibitor-containing highly active antiretroviral therapy.
J Acquir Immune Defic Syndr 2002; 29: 441–49.
140 Holme PA, Müller F, Solum NO, Brosstad F, Frøland SS,
Aukrust P. Enhanced activation of platelets with abnormal release
of RANTES in human immunodeficiency virus type 1 infection.
FASEB J 1998; 12: 79–89.
141 Mayne E, Funderburg NT, Sieg SF, et al. Increased platelet and
microparticle activation in HIV infection: upregulation of
P-selectin and tissue factor expression.
J Acquir Immune Defic Syndr 2012; 59: 340–46.
142 Funderburg NT, Mayne E, Sieg SF, et al. Increased tissue factor
expression on circulating monocytes in chronic HIV infection:
relationship to in vivo coagulation and immune activation. Blood
2010; 115: 161–67.
143 Lafeuillade A, Alessi MC, Poizot-Martin I, et al. Endothelial cell
dysfunction in HIV infection. J Acquir Immune Defic Syndr 1992;
5: 127–31.
144 Ross AC, Armentrout R, O’Riordan MA, et al. Endothelial
activation markers are linked to HIV status and are independent
of antiretroviral therapy and lipoatrophy.
J Acquir Immune Defic Syndr 2008; 49: 499–506.
145 Solages A, Vita JA, Thornton DJ, et al. Endothelial function in
HIV-infected persons. Clin Infect Dis 2006; 42: 1325–32.
146 Zietz C, Hotz B, Stürzl M, Rauch E, Penning R, Löhrs U. Aortic
endothelium in HIV-1 infection: chronic injury, activation, and
increased leukocyte adherence. Am J Pathol 1996; 149: 1887–98.
147 Joshi VV, Pawel B, Connor E, et al. Arteriopathy in children with
acquired immune deficiency syndrome. Pediatr Pathol 1987;
7: 261–75.
148 Mankhambo LA, Banda DL, Jeffers G, et al, and the IPD Study
Group. The role of angiogenic factors in predicting clinical
outcome in severe bacterial infection in Malawian children.
Crit Care Med 2010; 14: R91.
149 de Larrañaga GF, Bocassi AR, Puga LM, Alonso BS, Benetucci JA.
Endothelial markers and HIV infection in the era of highly active
antiretroviral treatment. Thromb Res 2003; 110: 93–98.
150 de Gaetano Donati K, Rabagliati R, Tumbarello M, et al. Increased
soluble markers of endothelial dysfunction in HIV-positive
patients under highly active antiretroviral therapy. AIDS 2003;
17: 765–68.
151 Rittirsch D, Redl H, Huber-Lang M. Role of complement in
multiorgan failure. Clin Dev Immunol 2012; 2012: 962927.
152 Huber G, Bánki Z, Lengauer S, Stoiber H. Emerging role for
complement in HIV infection. Curr Opin HIV AIDS 2011; 6: 419–26.
153 Yefenof E, Asjö B, Klein E. Alternative complement pathway
activation by HIV infected cells: C3 fixation does not lead to
complement lysis but enhances NK sensitivity. Int Immunol 1991;
3: 395–401.
108 www.thelancet.com/infection Vol 15 January 2015
Review
154 Dellinger RP, Levy MM, Rhodes A, et al, and the Surviving Sepsis
Campaign Guidelines Committee including the Pediatric
Subgroup. Surviving sepsis campaign: international guidelines
for management of severe sepsis and septic shock: 2012.
Crit Care Med 2013; 41: 580–637.
155 Hutchins NA, Unsinger J, Hotchkiss RS, Ayala A. The new
normal: immunomodulatory agents against sepsis immune
suppression. Trends Mol Med 2014; 20: 224–33.
156 Fagard C, Le Braz M, Günthard H, et al, and the Swiss HIV
Cohort Study. A controlled trial of granulocyte macrophage-colony
stimulating factor during interruption of HAART. AIDS 2003;
17: 1487–92.
157 Porichis F, Hart MG, Zupkosky J, et al. Differential impact of PD-
1 and/or interleukin-10 blockade on HIV-1-specific CD4 T cell and
antigen-presenting cell functions. J Virol 2014; 88: 2508–18.
158 Vanham G, Van Gulck E. Can immunotherapy be useful as a
“functional cure” for infection with Human Immunodeficiency
Virus-1? Retrovirology 2012; 9: 72.
159 Zhou H, Ji X, Wu Y, et al. A dual-role of Gu-4 in suppressing
HMGB1 secretion and blocking HMGB1 pro-inflammatory
activity during inflammation. PLoS one 2014; 9: e89634.
160 Vincent JL, Ramesh MK, Ernest D, et al. A randomized, double-
blind, placebo-controlled, Phase 2b study to evaluate the safety
and efficacy of recombinant human soluble thrombomodulin,
ART-123, in patients with sepsis and suspected disseminated
intravascular coagulation. Crit Care Med 2013; 41: 2069–79.

More Related Content

What's hot

1 infectious diseases
1 infectious diseases1 infectious diseases
1 infectious diseases
Engidaw Ambelu
 
Human genetic susceptibility to mycobacterium tuberculosis 1
Human genetic susceptibility to mycobacterium tuberculosis 1Human genetic susceptibility to mycobacterium tuberculosis 1
Human genetic susceptibility to mycobacterium tuberculosis 1
Shweta Kaul
 
Evasion of the host immune response by Mycobacterium tuberculosis
Evasion of the host immune response by Mycobacterium tuberculosisEvasion of the host immune response by Mycobacterium tuberculosis
Evasion of the host immune response by Mycobacterium tuberculosis
Richard Bautista
 
tuberculosis ppt by laxmi prasanna vemireddy
tuberculosis ppt by laxmi prasanna vemireddytuberculosis ppt by laxmi prasanna vemireddy
tuberculosis ppt by laxmi prasanna vemireddy
Laxmi Prasanna Vemireddy
 
Pathogenesis of microbial infections dr. ihsan alsaimary
Pathogenesis of microbial infections dr. ihsan alsaimaryPathogenesis of microbial infections dr. ihsan alsaimary
Pathogenesis of microbial infections dr. ihsan alsaimary
dr.Ihsan alsaimary
 
Infectious Disease
Infectious DiseaseInfectious Disease
Infectious Disease
Deep Deep
 
Immunology and Detection of Acute HIV-1 Infection
Immunology and Detection of Acute HIV-1 InfectionImmunology and Detection of Acute HIV-1 Infection
Immunology and Detection of Acute HIV-1 InfectionRongpong Plongla
 
Pulmmonary tuberculosis
Pulmmonary tuberculosisPulmmonary tuberculosis
Pulmmonary tuberculosisErasto Umoja
 
Infectious diseases
Infectious diseases Infectious diseases
Infectious diseases
Protik Biswas
 
Disease and infection
Disease and infectionDisease and infection
Disease and infection
Dr Ifsha Akhlaq
 
infections and immunity
infections and immunityinfections and immunity
infections and immunity
Ashish Jawarkar
 
Ppat.1005667
Ppat.1005667Ppat.1005667
Ppat.1005667
Julio A. Diaz M.
 
Infectious Diseases
Infectious DiseasesInfectious Diseases
Infectious Diseases
OMICS International
 
Hypersensitivity Reactions
Hypersensitivity ReactionsHypersensitivity Reactions
Hypersensitivity Reactions
Anantha Chaitanya
 

What's hot (20)

1 infectious diseases
1 infectious diseases1 infectious diseases
1 infectious diseases
 
Human genetic susceptibility to mycobacterium tuberculosis 1
Human genetic susceptibility to mycobacterium tuberculosis 1Human genetic susceptibility to mycobacterium tuberculosis 1
Human genetic susceptibility to mycobacterium tuberculosis 1
 
Immunopathology 2
Immunopathology 2Immunopathology 2
Immunopathology 2
 
Evasion of the host immune response by Mycobacterium tuberculosis
Evasion of the host immune response by Mycobacterium tuberculosisEvasion of the host immune response by Mycobacterium tuberculosis
Evasion of the host immune response by Mycobacterium tuberculosis
 
Tuberclosis
TuberclosisTuberclosis
Tuberclosis
 
Immunodeficiencies
ImmunodeficienciesImmunodeficiencies
Immunodeficiencies
 
tuberculosis ppt by laxmi prasanna vemireddy
tuberculosis ppt by laxmi prasanna vemireddytuberculosis ppt by laxmi prasanna vemireddy
tuberculosis ppt by laxmi prasanna vemireddy
 
Pathogenesis of microbial infections dr. ihsan alsaimary
Pathogenesis of microbial infections dr. ihsan alsaimaryPathogenesis of microbial infections dr. ihsan alsaimary
Pathogenesis of microbial infections dr. ihsan alsaimary
 
Infectious Part 1
Infectious Part 1Infectious Part 1
Infectious Part 1
 
Infectious Disease
Infectious DiseaseInfectious Disease
Infectious Disease
 
Immunology and Detection of Acute HIV-1 Infection
Immunology and Detection of Acute HIV-1 InfectionImmunology and Detection of Acute HIV-1 Infection
Immunology and Detection of Acute HIV-1 Infection
 
Pulmmonary tuberculosis
Pulmmonary tuberculosisPulmmonary tuberculosis
Pulmmonary tuberculosis
 
Infectious diseases
Infectious diseases Infectious diseases
Infectious diseases
 
Disease and infection
Disease and infectionDisease and infection
Disease and infection
 
hd Subbarao
hd Subbaraohd Subbarao
hd Subbarao
 
infections and immunity
infections and immunityinfections and immunity
infections and immunity
 
Ppat.1005667
Ppat.1005667Ppat.1005667
Ppat.1005667
 
Infectious Diseases
Infectious DiseasesInfectious Diseases
Infectious Diseases
 
McAlister_VME 158 Paper
McAlister_VME 158 Paper McAlister_VME 158 Paper
McAlister_VME 158 Paper
 
Hypersensitivity Reactions
Hypersensitivity ReactionsHypersensitivity Reactions
Hypersensitivity Reactions
 

Similar to Vih y paciente septico

joURNAL READING BIOMARKER.pptx
joURNAL READING BIOMARKER.pptxjoURNAL READING BIOMARKER.pptx
joURNAL READING BIOMARKER.pptx
joganks1
 
AIDS PRESANTATION.pptx
AIDS PRESANTATION.pptxAIDS PRESANTATION.pptx
AIDS PRESANTATION.pptx
Mkindi Mkindi
 
HIV and Lung health
HIV and Lung healthHIV and Lung health
HIV and Lung health
Arnab Nandy
 
CNS IRIS.pptx
CNS IRIS.pptxCNS IRIS.pptx
CNS IRIS.pptx
NeurologyKota
 
CNS IRIS.pptx
CNS IRIS.pptxCNS IRIS.pptx
CNS IRIS.pptx
vijay prakash
 
immunobiology of hiv virus human immunodeficeincy virus
immunobiology of hiv virus human immunodeficeincy virusimmunobiology of hiv virus human immunodeficeincy virus
immunobiology of hiv virus human immunodeficeincy virus
Akshay Raj
 
The role of infections in autoimmune disease
The role of infections in autoimmune diseaseThe role of infections in autoimmune disease
The role of infections in autoimmune diseaseLily Fernandez Mullins
 
Rheumatic Fever 2023 May.pdf
Rheumatic  Fever 2023 May.pdfRheumatic  Fever 2023 May.pdf
Rheumatic Fever 2023 May.pdf
JaiPrakashSoni8
 
Infectious Diseases 12
Infectious Diseases  12Infectious Diseases  12
Infectious Diseases 12pgr224
 
Unit 16 HIV-AIDS (1).pptx
Unit 16 HIV-AIDS (1).pptxUnit 16 HIV-AIDS (1).pptx
Unit 16 HIV-AIDS (1).pptx
BarentuShemsu
 
ĐIỀU TRỊ ĐIỀU HÒA MIỄN DỊCH Ở BN COVID 19
ĐIỀU TRỊ ĐIỀU HÒA MIỄN DỊCH Ở BN COVID 19ĐIỀU TRỊ ĐIỀU HÒA MIỄN DỊCH Ở BN COVID 19
ĐIỀU TRỊ ĐIỀU HÒA MIỄN DỊCH Ở BN COVID 19
SoM
 
TUBERCULOSIS.pptx
TUBERCULOSIS.pptxTUBERCULOSIS.pptx
TUBERCULOSIS.pptx
DR MUKESH SAH
 
Human Immunodeficiency Virus
Human Immunodeficiency VirusHuman Immunodeficiency Virus
Human Immunodeficiency Viruspromotemedical
 
1-s2.0-S1472979215302419-main
1-s2.0-S1472979215302419-main1-s2.0-S1472979215302419-main
1-s2.0-S1472979215302419-mainKarina Chavez
 
Acquired immunodeficiency syndrome.pptx
Acquired immunodeficiency  syndrome.pptxAcquired immunodeficiency  syndrome.pptx
Acquired immunodeficiency syndrome.pptx
The Microbiology Meducator
 
Infectious diseases
Infectious diseasesInfectious diseases
Infectious diseases
ERIC GENERAL
 
patho3.pptx
patho3.pptxpatho3.pptx
patho3.pptx
Sani191640
 
Immune responses to infectious diseases
Immune responses to infectious diseases Immune responses to infectious diseases
Immune responses to infectious diseases
Hadia Azhar
 
Rheumatic feve 2018
Rheumatic feve  2018Rheumatic feve  2018
Rheumatic feve 2018
rajasthan govt
 
Mecanismo les
Mecanismo lesMecanismo les
Mecanismo les
Joel Choca
 

Similar to Vih y paciente septico (20)

joURNAL READING BIOMARKER.pptx
joURNAL READING BIOMARKER.pptxjoURNAL READING BIOMARKER.pptx
joURNAL READING BIOMARKER.pptx
 
AIDS PRESANTATION.pptx
AIDS PRESANTATION.pptxAIDS PRESANTATION.pptx
AIDS PRESANTATION.pptx
 
HIV and Lung health
HIV and Lung healthHIV and Lung health
HIV and Lung health
 
CNS IRIS.pptx
CNS IRIS.pptxCNS IRIS.pptx
CNS IRIS.pptx
 
CNS IRIS.pptx
CNS IRIS.pptxCNS IRIS.pptx
CNS IRIS.pptx
 
immunobiology of hiv virus human immunodeficeincy virus
immunobiology of hiv virus human immunodeficeincy virusimmunobiology of hiv virus human immunodeficeincy virus
immunobiology of hiv virus human immunodeficeincy virus
 
The role of infections in autoimmune disease
The role of infections in autoimmune diseaseThe role of infections in autoimmune disease
The role of infections in autoimmune disease
 
Rheumatic Fever 2023 May.pdf
Rheumatic  Fever 2023 May.pdfRheumatic  Fever 2023 May.pdf
Rheumatic Fever 2023 May.pdf
 
Infectious Diseases 12
Infectious Diseases  12Infectious Diseases  12
Infectious Diseases 12
 
Unit 16 HIV-AIDS (1).pptx
Unit 16 HIV-AIDS (1).pptxUnit 16 HIV-AIDS (1).pptx
Unit 16 HIV-AIDS (1).pptx
 
ĐIỀU TRỊ ĐIỀU HÒA MIỄN DỊCH Ở BN COVID 19
ĐIỀU TRỊ ĐIỀU HÒA MIỄN DỊCH Ở BN COVID 19ĐIỀU TRỊ ĐIỀU HÒA MIỄN DỊCH Ở BN COVID 19
ĐIỀU TRỊ ĐIỀU HÒA MIỄN DỊCH Ở BN COVID 19
 
TUBERCULOSIS.pptx
TUBERCULOSIS.pptxTUBERCULOSIS.pptx
TUBERCULOSIS.pptx
 
Human Immunodeficiency Virus
Human Immunodeficiency VirusHuman Immunodeficiency Virus
Human Immunodeficiency Virus
 
1-s2.0-S1472979215302419-main
1-s2.0-S1472979215302419-main1-s2.0-S1472979215302419-main
1-s2.0-S1472979215302419-main
 
Acquired immunodeficiency syndrome.pptx
Acquired immunodeficiency  syndrome.pptxAcquired immunodeficiency  syndrome.pptx
Acquired immunodeficiency syndrome.pptx
 
Infectious diseases
Infectious diseasesInfectious diseases
Infectious diseases
 
patho3.pptx
patho3.pptxpatho3.pptx
patho3.pptx
 
Immune responses to infectious diseases
Immune responses to infectious diseases Immune responses to infectious diseases
Immune responses to infectious diseases
 
Rheumatic feve 2018
Rheumatic feve  2018Rheumatic feve  2018
Rheumatic feve 2018
 
Mecanismo les
Mecanismo lesMecanismo les
Mecanismo les
 

More from Aivan Lima

lra.pptx
lra.pptxlra.pptx
lra.pptx
Aivan Lima
 
Anestesia
AnestesiaAnestesia
Anestesia
Aivan Lima
 
Neuro
NeuroNeuro
Neuro
Aivan Lima
 
Renal
RenalRenal
Renal
Aivan Lima
 
Ventilación Mecánica en la paciente obstétrica
Ventilación Mecánica en la paciente obstétricaVentilación Mecánica en la paciente obstétrica
Ventilación Mecánica en la paciente obstétrica
Aivan Lima
 
Hipertensivas
HipertensivasHipertensivas
Hipertensivas
Aivan Lima
 
Manejo inicial gran quemado ii
Manejo inicial gran quemado iiManejo inicial gran quemado ii
Manejo inicial gran quemado ii
Aivan Lima
 
Manejo inicial gran quemado i
Manejo inicial gran quemado iManejo inicial gran quemado i
Manejo inicial gran quemado i
Aivan Lima
 
Manejo ambulatorio de quemaduras
Manejo ambulatorio de quemadurasManejo ambulatorio de quemaduras
Manejo ambulatorio de quemaduras
Aivan Lima
 
Ngal lra
Ngal lraNgal lra
Ngal lra
Aivan Lima
 
Lra que hay de nuevo
Lra que hay de nuevoLra que hay de nuevo
Lra que hay de nuevo
Aivan Lima
 
Advances aki
Advances akiAdvances aki
Advances aki
Aivan Lima
 
Lra lancet
Lra lancetLra lancet
Lra lancet
Aivan Lima
 
Nice sugar nejm 2009
Nice sugar nejm 2009Nice sugar nejm 2009
Nice sugar nejm 2009
Aivan Lima
 
Hiperglucemia en uci 2009 med intensiva
Hiperglucemia en uci 2009 med intensivaHiperglucemia en uci 2009 med intensiva
Hiperglucemia en uci 2009 med intensiva
Aivan Lima
 
Tratamiento del choque septico
Tratamiento del choque septicoTratamiento del choque septico
Tratamiento del choque septico
Aivan Lima
 
Liquidos en el paciente septico
Liquidos en el paciente septicoLiquidos en el paciente septico
Liquidos en el paciente septico
Aivan Lima
 
Sepsis primer
Sepsis primerSepsis primer
Sepsis primer
Aivan Lima
 
The japanese guidelines fot the management of sepsis j int care 2014
The japanese guidelines fot the management of sepsis j int care 2014The japanese guidelines fot the management of sepsis j int care 2014
The japanese guidelines fot the management of sepsis j int care 2014
Aivan Lima
 
Surviving sepsis guidelines
Surviving sepsis guidelinesSurviving sepsis guidelines
Surviving sepsis guidelines
Aivan Lima
 

More from Aivan Lima (20)

lra.pptx
lra.pptxlra.pptx
lra.pptx
 
Anestesia
AnestesiaAnestesia
Anestesia
 
Neuro
NeuroNeuro
Neuro
 
Renal
RenalRenal
Renal
 
Ventilación Mecánica en la paciente obstétrica
Ventilación Mecánica en la paciente obstétricaVentilación Mecánica en la paciente obstétrica
Ventilación Mecánica en la paciente obstétrica
 
Hipertensivas
HipertensivasHipertensivas
Hipertensivas
 
Manejo inicial gran quemado ii
Manejo inicial gran quemado iiManejo inicial gran quemado ii
Manejo inicial gran quemado ii
 
Manejo inicial gran quemado i
Manejo inicial gran quemado iManejo inicial gran quemado i
Manejo inicial gran quemado i
 
Manejo ambulatorio de quemaduras
Manejo ambulatorio de quemadurasManejo ambulatorio de quemaduras
Manejo ambulatorio de quemaduras
 
Ngal lra
Ngal lraNgal lra
Ngal lra
 
Lra que hay de nuevo
Lra que hay de nuevoLra que hay de nuevo
Lra que hay de nuevo
 
Advances aki
Advances akiAdvances aki
Advances aki
 
Lra lancet
Lra lancetLra lancet
Lra lancet
 
Nice sugar nejm 2009
Nice sugar nejm 2009Nice sugar nejm 2009
Nice sugar nejm 2009
 
Hiperglucemia en uci 2009 med intensiva
Hiperglucemia en uci 2009 med intensivaHiperglucemia en uci 2009 med intensiva
Hiperglucemia en uci 2009 med intensiva
 
Tratamiento del choque septico
Tratamiento del choque septicoTratamiento del choque septico
Tratamiento del choque septico
 
Liquidos en el paciente septico
Liquidos en el paciente septicoLiquidos en el paciente septico
Liquidos en el paciente septico
 
Sepsis primer
Sepsis primerSepsis primer
Sepsis primer
 
The japanese guidelines fot the management of sepsis j int care 2014
The japanese guidelines fot the management of sepsis j int care 2014The japanese guidelines fot the management of sepsis j int care 2014
The japanese guidelines fot the management of sepsis j int care 2014
 
Surviving sepsis guidelines
Surviving sepsis guidelinesSurviving sepsis guidelines
Surviving sepsis guidelines
 

Recently uploaded

BRACHYTHERAPY OVERVIEW AND APPLICATORS
BRACHYTHERAPY OVERVIEW  AND  APPLICATORSBRACHYTHERAPY OVERVIEW  AND  APPLICATORS
BRACHYTHERAPY OVERVIEW AND APPLICATORS
Krishan Murari
 
THOA 2.ppt Human Organ Transplantation Act
THOA 2.ppt Human Organ Transplantation ActTHOA 2.ppt Human Organ Transplantation Act
THOA 2.ppt Human Organ Transplantation Act
DrSathishMS1
 
ANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptxANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptx
Swetaba Besh
 
How to Give Better Lectures: Some Tips for Doctors
How to Give Better Lectures: Some Tips for DoctorsHow to Give Better Lectures: Some Tips for Doctors
How to Give Better Lectures: Some Tips for Doctors
LanceCatedral
 
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model SafeSurat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Savita Shen $i11
 
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
GL Anaacs
 
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 UpakalpaniyaadhyayaCharaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Dr KHALID B.M
 
MANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdf
MANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdfMANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdf
MANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdf
Jim Jacob Roy
 
Evaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animalsEvaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animals
Shweta
 
New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...
New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...
New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...
i3 Health
 
Are There Any Natural Remedies To Treat Syphilis.pdf
Are There Any Natural Remedies To Treat Syphilis.pdfAre There Any Natural Remedies To Treat Syphilis.pdf
Are There Any Natural Remedies To Treat Syphilis.pdf
Little Cross Family Clinic
 
Antiulcer drugs Advance Pharmacology .pptx
Antiulcer drugs Advance Pharmacology .pptxAntiulcer drugs Advance Pharmacology .pptx
Antiulcer drugs Advance Pharmacology .pptx
Rohit chaurpagar
 
Physiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of TastePhysiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of Taste
MedicoseAcademics
 
Superficial & Deep Fascia of the NECK.pptx
Superficial & Deep Fascia of the NECK.pptxSuperficial & Deep Fascia of the NECK.pptx
Superficial & Deep Fascia of the NECK.pptx
Dr. Rabia Inam Gandapore
 
Non-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdfNon-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdf
MedicoseAcademics
 
Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...
Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...
Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...
Savita Shen $i11
 
ARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTS
ARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTSARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTS
ARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTS
Dr. Vinay Pareek
 
heat stroke and heat exhaustion in children
heat stroke and heat exhaustion in childrenheat stroke and heat exhaustion in children
heat stroke and heat exhaustion in children
SumeraAhmad5
 
basicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdfbasicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdf
aljamhori teaching hospital
 
Surgical Site Infections, pathophysiology, and prevention.pptx
Surgical Site Infections, pathophysiology, and prevention.pptxSurgical Site Infections, pathophysiology, and prevention.pptx
Surgical Site Infections, pathophysiology, and prevention.pptx
jval Landero
 

Recently uploaded (20)

BRACHYTHERAPY OVERVIEW AND APPLICATORS
BRACHYTHERAPY OVERVIEW  AND  APPLICATORSBRACHYTHERAPY OVERVIEW  AND  APPLICATORS
BRACHYTHERAPY OVERVIEW AND APPLICATORS
 
THOA 2.ppt Human Organ Transplantation Act
THOA 2.ppt Human Organ Transplantation ActTHOA 2.ppt Human Organ Transplantation Act
THOA 2.ppt Human Organ Transplantation Act
 
ANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptxANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptx
ANATOMY AND PHYSIOLOGY OF URINARY SYSTEM.pptx
 
How to Give Better Lectures: Some Tips for Doctors
How to Give Better Lectures: Some Tips for DoctorsHow to Give Better Lectures: Some Tips for Doctors
How to Give Better Lectures: Some Tips for Doctors
 
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model SafeSurat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
 
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
HOT NEW PRODUCT! BIG SALES FAST SHIPPING NOW FROM CHINA!! EU KU DB BK substit...
 
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 UpakalpaniyaadhyayaCharaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
Charaka Samhita Sutra sthana Chapter 15 Upakalpaniyaadhyaya
 
MANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdf
MANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdfMANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdf
MANAGEMENT OF ATRIOVENTRICULAR CONDUCTION BLOCK.pdf
 
Evaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animalsEvaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animals
 
New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...
New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...
New Directions in Targeted Therapeutic Approaches for Older Adults With Mantl...
 
Are There Any Natural Remedies To Treat Syphilis.pdf
Are There Any Natural Remedies To Treat Syphilis.pdfAre There Any Natural Remedies To Treat Syphilis.pdf
Are There Any Natural Remedies To Treat Syphilis.pdf
 
Antiulcer drugs Advance Pharmacology .pptx
Antiulcer drugs Advance Pharmacology .pptxAntiulcer drugs Advance Pharmacology .pptx
Antiulcer drugs Advance Pharmacology .pptx
 
Physiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of TastePhysiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of Taste
 
Superficial & Deep Fascia of the NECK.pptx
Superficial & Deep Fascia of the NECK.pptxSuperficial & Deep Fascia of the NECK.pptx
Superficial & Deep Fascia of the NECK.pptx
 
Non-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdfNon-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdf
 
Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...
Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...
Phone Us ❤85270-49040❤ #ℂall #gIRLS In Surat By Surat @ℂall @Girls Hotel With...
 
ARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTS
ARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTSARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTS
ARTHROLOGY PPT NCISM SYLLABUS AYURVEDA STUDENTS
 
heat stroke and heat exhaustion in children
heat stroke and heat exhaustion in childrenheat stroke and heat exhaustion in children
heat stroke and heat exhaustion in children
 
basicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdfbasicmodesofventilation2022-220313203758.pdf
basicmodesofventilation2022-220313203758.pdf
 
Surgical Site Infections, pathophysiology, and prevention.pptx
Surgical Site Infections, pathophysiology, and prevention.pptxSurgical Site Infections, pathophysiology, and prevention.pptx
Surgical Site Infections, pathophysiology, and prevention.pptx
 

Vih y paciente septico

  • 1. www.thelancet.com/infection Vol 15 January 2015 95 Review The effect of HIV infection on the host response to bacterial sepsis Michaëla A M Huson, Martin P Grobusch, Tom van der Poll Bacterial sepsis is an important cause of morbidity and mortality in patients with HIV. HIV causes increased susceptibility to invasive infections and affects sepsis pathogenesis caused by pre-existing activation and exhaustion of the immune system. We review the effect of HIV on different components of immune responses implicated in bacterial sepsis, and possible mechanisms underlying the increased risk of invasive bacterial infections. We focus on pattern recognition receptors and innate cellular responses, cytokines, lymphocytes, coagulation, and the complement system. A combination of factors causes increased susceptibility to infection and can contribute to a disturbed immune response during a septic event in patients with HIV. HIV-induced perturbations of the immune system depend on stage of infection and are only in part restored by combination antiretroviral therapy. Immuno- modulatory treatments currently under development for sepsis might be particularly beneficial to patients with HIV co-infection because many pathogenic mechanisms in HIV and sepsis overlap. Introduction People infected with HIV are at increased risk of developing other infections. Reports from developed countries show that, with the introduction of combination antiretroviral therapy (cART), the incidence of opportunistic infections such as Pneumocystis jirovecii pneumonia has decreased substantially, and sepsis is now the principal cause of intensive care unit (ICU) admission and death in patients with HIV who are admitted to hospital.1 Worldwide, patients with HIV are at increased risk of developing bacterial bloodstream infections, particularly with non-typhoidal salmonella (NTS) and Streptococcus pneumoniae,2,3 even in those using effective cART.4 In developing regions with high rates of HIV infection, the scale of the problem is immense. In African hospitals, 31–83% of patients presenting with fever are infected with HIV, and 10–32% of these patients have bacterial bloodstream infections, with mortality rates of 25–46% (table 1).5-15 However, little is known about the particularities of the host response during bacterial sepsis in patients with HIV. The immune response during sepsis is thought to be an imbalance between proinflammatory and anti- inflammatory effects, which result in organ failure.16,17 Likewise, HIV infection is characterised by a combination of immune suppression and chronic inflammation, which results in exhaustion of the immune system.18 In this Review we describe how different components of the immune response might be affected by HIV during bacterial sepsis, and possible mechanisms for the increased risk of invasive bacterial infections. We also discuss parallels in immuno- modulatory therapies proposed for sepsis and HIV, which represent an appealing area for future research. First line defences Overview A sepsis event typically starts with a pathogen invading a normally sterile site. The first line of defence consists of epithelial cells of the skin, gut, and lungs. After breaching the epithelial barrier, pathogens are first detected by innate immune cells, including monocytes, macrophages, dendritic cells, and neutrophils.19 These cells express pattern recognition receptors (PRRs) on their surface and in the cytosol to enable recognition of conserved motifs expressed by pathogens.20,21 Four classes of PRRs have been identified: toll-like receptors (TLRs), C-type lectin receptors (CLRs), nucleotide- binding oligomerisation domain leucine-rich-repeat containing receptors (NOD-LRRs), and retinoic acid- inducible gene I protein helicase receptors (RLRs).20 In the context of sepsis pathogenesis, TLRs have been studied the most extensively. They are expressed on the cell surface (TLRs 1, 2, 4, 5, and 6) to enable recognition of bacterial outer membrane components, and in intracellular compartments (TLRs 3, 7, 8, and 9) for detection of nucleic acids derived from intracellular pathogens, mainly viruses.20 For bacterial recognition, TLRs 2, 4, 5, and 9 are the most important. Additionally, TLR7 can sense bacterial RNA released within phagosomal vacuoles,22 and TLR3 can function as an amplifier of host RNA-triggered inflammation during sepsis.23 Ligand recognition by TLRs triggers a signalling cascade, which results in the production of cytokines.20 Although essential for pathogen recognition and the innate immune response, uncontrolled stimulation causes excessive inflammation; hence TLR signalling is normally tightly regulated. Innate immune cells have several specific functions in antibacterial immunity. Monocytes and macrophages have a crucial role in maintaining homoeostasis by phagocytosis of apoptotic cells and microorganisms.19 As the main producers of proinflammatory cytokines, they are also thought to be key in sepsis pathogenesis.19 Dendritic cells are the main antigen-presenting cells; maturation is induced after ingestion of antigen, followed by migration to lymphoid tissue. Mature dendritic cells express co-stimulatory molecules on their surface, which synergise with antigen to activate T cells.24 Natural killer (NK) cells are implicated in Lancet Infect Dis 2015; 15: 95–108 Published Online October 20, 2014 http://dx.doi.org/10.1016/ S1473-3099(14)70917-X Division of Infectious Diseases, Centre of Experimental and Molecular Medicine (M A M Huson MD, ProfT van der Poll MD); and Division of Infectious Diseases, Centre ofTropical Medicine and Travel Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands (Prof M P Grobusch MD) Correspondence to: Dr Michaëla A M Huson, Academic Medical Centre, University of Amsterdam, Amsterdam 1105 AZ, Netherlands m.a.huson@amc.uva.nl
  • 2. 96 www.thelancet.com/infection Vol 15 January 2015 Review antibacterial immune responses through the ability to directly lyse infected cells, provide early sources of proinflammatory cytokines, predominantly inter- feron-γ, induce dendritic cell maturation, and amplify the proinflammatory effects of myeloid cells.25,26 Likewise, natural killer T cells (NK T cells), a subset that shares cell-surface proteins with conventional T cells and NK cells, have been implicated in sepsis pathogenesis because of their strong proinflammatory cytokine release.27 Recruited neutrophils form an additional important first line of defence against invading pathogens. They kill microbes through phagocytosis, the release of lytic enzymes from their granules, the production of reactive oxygen inter- mediates, and the formation of neutrophil extracellular traps (NETs)—lattices of chromatin decorated with anti- microbial proteins with strong bactericidal capacity.28 The innate immune system is thought to be mostly responsible for the excessive release of proinflammatory cytokines early in sepsis pathogenesis.29 The most extensively studied proinflammatory cytokines in sepsis are tumour necrosis factor α (TNFα) and interleukin 1β, both of which are capable of activating target cells and induce the production of more inflammatory mediators.29 Additionally, host cells release damage-associated molecular patterns (DAMPs) in response to pathogens or injury, which are recognised by PRRs, thereby enhancing immune activation. The most investigated DAMP is HMGB1, which signals via TLR2, TLR4, and TLR9 to induce cytokine release, activation of coagulation, and neutrophil recruitment.30 Proinflammatory cytokines and DAMPs seem to have a double role in sepsis pathogenesis; they are essential for an adequate innate defence during early stage infection, but also contribute to hyperinflammation during late phase uncontrolled infection.20,21 In patients with severe sepsis and in murine sepsis models, high concentrations of interleukins 1, 6, 8, and 17, CCL2, CSF3, and HMGB1 are associated with mortality.29 HIV affects the first lines of defence in several ways. First, defects of epithelial barriers are common and have been particularly well described for the gut. In the gut, HIV causes barrier defects during acute infection, which are maintained during chronic infection, thereby enabling invasive infections by intestinal pathogens such as non-typhoidal salmonella.31 Furthermore, microbial products that translocate into the circulation fuel chronic immune activation and exhaustion.32 Toll-like receptors HIV infection and AIDS are associated with increased TLR2, TLR3, TLR4, TLR7, and TLR9 expression on various cells, including T lymphocytes, monocytes, macrophages, and dendritic cells,33–38 although one study34 reported lower peripheral blood mononuclear cell (PBMC) RNA expression levels of TLR3, TLR4, and TLR9 in patients with chronic HIV unresponsive to cART. Reports on the functional effect of differential TLR expression are inconsistent. Ex-vivo studies with PBMCs from patients with HIV noted a correlation between increased expression of TLRs and increased TNFα production after stimulation with lipopolysaccharide, a Study location andtimeframe Primary inclusion criteria Patients infectedwith HIV (%ofpatients tested) CA bacterial BSI in patients with HIV (%) Main isolates in patientswith HIV (%) Mortality in patients with HIV withCA bacterial BSI Archibald, 19985 UrbanTanzania, 1995 Febrile (≥37·5°C) admission 282 (55%) 51 (18%) NTS (45%), Escherichiacoli (14%), Streptococcus pneumoniae (12%) Not reported Archibald, 20006 Urban Malawi, 1997 Febrile (≥37·5°C) admissions 173 (74%) 37 (21%) Spneumoniae (57%), NTS (30%) Not reported Arthur, 20017 Urban Kenya, 1988–97 Hospital admission 436 (32%) 87 (20%) NTS (46%), Spneumoniae (33%), Ecoli (6%) 39% Bell, 20018 Urban Malawi, 1998 Febrile (37·5°C) admissions 173 (73%) 36 (21%) NTS (62%), Ecoli (7%), Salmonella Typhi (7%) Not reported Crump, 20119 RuralTanzania, 2007–08 Febrile (≥38°C) admissions 161 (40%) 26 (16%) Spneumoniae (54%), Ecoli (12%), NTS (8%), STyphi (8%) Not reported Grant, 199710 Ivory coast, 1995 Admission tothe infectious diseaseunit 198 (79%) 39 (20%) NTS (59%), Ecoli (15%), Spneumoniae (10%) 46% Mayanja, 201011 RuralUganda, 1996–2007 Fever (≥38°C)with no detectable malaria parasites 488 (64%) 152 (31%) Spneumoniae (43%), NTS (26%), Ecoli (6%) Not reported Meremo, 201212 UrbanTanzania, 2011 Hospital admission with fever (>37·5°C) 156 (45%) 16 (10%) NTS (75%) 35% Nadjm, 201213 RuralTanzania, 2007 Fever and one severity criterion 69 (35%) 12 (17%) NTS (25%), Spneumoniae (25%), Streptococcus pyogenes (17%) 25% Peters, 200414 Urban Malawi, 2000 Admission with fever (≥37·4°C)or historyof fever inthe past 4days 291 (83%) 66 (23%) NTS (67%), Spneumoniae (20%), Ecoli (6%) Not reported Ssali, 199815 Uganda, 1997 Admission with fever (>38°C) 222 (76%) 33 (15%) Salmonella spp (39%), Spneumoniae (33%) Not reported Data was derived from prospective observational studies, which were previously described by Huson and colleagues.3 CA=community acquired. BSI=bloodstream infection. NTS=non-typhoidal salmonella. Table 1: Burden of community-acquired bacterial bloodstream infections in patients with HIV in African countries where HIV is highly prevalent
  • 3. www.thelancet.com/infection Vol 15 January 2015 97 Review See Online for appendix TLR4 ligand.36,38 By contrast, alveolar macrophages from patients with HIV showed a classic activation phenotype with increased TLR4 expression, although binding, internalisation, and killing of opsonised S pneumoniae bacteria, which signal via TLR2, TLR4, and TLR9, were similar to HIV-negative controls.37 TLR3 and TLR7 are known to be key modulators in anti-HIV immunity, and HIV-induced changes in expression of these TLRs might have a role during antibacterial defence, but no studies have yet investigated this subject. Most evidence suggests that upregulation of TLRs is responsible for cell activation in response to bacteria in patients with HIV, especially in those with advanced disease, although functional consequences differ according to cell type. Increased expression of TLRs on cytokine-producing cells might contribute to aberrant inflammation in the disturbed homoeostasis in sepsis, although this possibility has not yet been addressed in in-vivo studies. Monocytes and macrophages The effect of HIV on monocytes and macrophages is mainly indirect because only a small percentage of these cells are infected with HIV.39 HIV-mediated defects in phagocytosis, cell signalling, and cytokine production have been described, although results vary between studies (appendix). Ex-vivo studies using monocytes or monocyte-derived macrophages from patients with HIV, reported reduced phagocytosis of Escherichia coli40 and, in symptomatic patients with HIV, Staphylococcus aureus.41 By contrast, others observed normal phagocytic function of macrophages towards opsonised E coli and S aureus in vitro,42 and increased monocyte phagocytosis of these pathogens in cells harvested from patients with early HIV infection.43 In peripheral blood, both monocytes and macrophages appear to show an HIV-induced, primed, proinflammatory state, with increased cytokine release on stimulation with TLR ligands.44,45 A non-classic subset of monocytes with high expression of M-DC8, a subset that is also increased in inflamed tissue in chronic inflammatory diseases such as rheumatoid arthritis, was identified as the predominant cell type responsible for TNFα overproduction46 (table 2).47–67 Several studies also examined the effect of HIV on the alveolar compartment. Although normal alveolar Study condition* HIV-induced changes Comments Whole blood In vivo ↑ Interleukin 6, interleukin 10,TNFα, interleukin 1α, interleukin 1β, interleukin 8, CCL2, interleukin 1R1, and interferon γ47,48 ↓ interleukin 12, andCSF248,49 Cytokine levels normalised aftertreatmentwith cART47,48 Two studies50,51 examined cytokine levels in sepsis patients, but recorded no differences, except higher interleukin 10 concentrations in HIV co-infected patients inoneofthese studies50 DecreasedCSF2 levelswere seen in patientswith HIVwith advanceddisease Peripheral blood mononuclear cells Ex-vivo and in- vitro stimulation ↑TNFα 38,46,52 ·· Monocytes Ex-vivo stimulation ↑ Interleukin 6, interleukin 1β,TNFα44 ·· M-DC8 monocytes Ex-vivo stimulation ↑TNFα46 These cellswere identified asthe main celltype responsible forTNFα overproduction46 Increased levelsof M-DC8 monocyteswere recorded in viraemic patients with HIV, but not in virally suppressed patientson cART46 Alveolar macrophages Ex-vivo stimulation ↓TNFα, interleukin 8, interleukin 1253–56 Reports conflictonthis subject;other studies noted increased interleukin 8, 12 and 10 in alveolar macrophagesof patientswith HIV56 Impaired interleukin 12 productionwasonly reported in patientswith advanceddisease56 Dendritic cells In-vitro and ex- vivo stimulation ↓ Interleukin 12,57–59 interleukin 6,58 interferon γ,γ,58,6058,60 interferon α,interferon α,59,6159,61 interleukin 2,interleukin 2,6161 and interleukin 10and interleukin 106060 Most reportsdescribedecreased cytokine production after stimulation, but normal,62 or even increased cytokine responsesto stimulationwere also reported63 Natural killer cells Ex-vivo stimulation ↓ Interferon γ64 The effectwasobserved in both cART-treated anduntreated patients64 Natural killer T cells Ex-vivo stimulation ↓ Interferon γ and interleukin 465 Stimulationsweredonewith marine sponge-derived α-galactosylceramide (αGalCer) as a ligand;65 similar studiesusing bacterial ligands as a stimulus were not available Cytokine secretionwas restored by cART65 CD4T cells In-vivo ↑ Interleukin 1066 Interleukin 10 productionwas increased in chronic progressors and recently infected individuals, but not in non-progressorswith HIV66 Gut mucosa Th17 cells Ex-vivo stimulation ↓ Interleukin 22,TNFα, and interferon γ67 ↑ Interleukin 1067 Restorationof cytokine responseswas seen after prolongedtreatmentwith cART67 cART=combination antiretroviral therapy. *All studies are in human beings or used human cells, or human cell lines; all stimulations were done with bacteria or bacterial products unless otherwise specified. Table 2: Effect of HIV infection on the cytokine milieu by cell type
  • 4. 98 www.thelancet.com/infection Vol 15 January 2015 Review macrophage phagocytosis of S pneumoniae was previously reported,37 a study68 noted impaired phago- cytosis in HIV-infected small alveolar macrophages, as well as impaired proteolysis of phagosomes in both infected and uninfected alveolar macrophages. Cytokine release, including that of interleukin 8, in response to S pneumoniae, and TNFα, in response to TLR2 and TLR4 ligands, was impaired in alveolar macrophages from patients with HIV (table 2).53–55 Considering the crucial role of TNFα in resisting experimental S pneumoniae infections in mice,69 and the established part in neutrophil recruitment to sites of infection by interleukin 8, a reduced cytokine response by alveolar macrophages might contribute to increased susceptibility to invasive pneumococcal disease in patients with HIV (figure 1). Another study56 noted increased production of both proinflammatory (interleukin 8 and 12) and anti-inflammatory (interleukin 10) cytokines by alveolar macrophages of asymptomatic patients with HIV after stimulation with S aureus. However, in patients with more advanced disease interleukin 12 release was reduced after stimulation, which was mediated by interleukin 10.56 Interleukin 12 is known to induce T-helper (Th) 1 development, which is essential for defence against intracellular pathogens, such as non-typhoidal salmonella, but also has a role in the immune response against S pneumoniae.70 Finally, alveolar macrophage apoptosis, a critical host cell response for control of infection, including pneumococcal pneumonia,71 might be impaired in patients with HIV.72 Most evidence suggests impairment of phagocytosis by macrophages in individuals with HIV, which is accompanied by impaired proinflammatory cytokine release, especially in alveolar macrophages. These defects might contribute to increased susceptibility to bacterial infection and deregulated inflammation. Dendritic cells In patients with HIV, around 10–15% of dendritic cells are infected,57 but HIV also affects dendritic cell function in indirect ways (appendix). Patients with HIV have reduced peripheral blood dendritic cell numbers,61,63,73,74 which are inversely correlated to viral ↑S pneumoniae ↓Interleukin 17 ↓CD4 T cells ↓Macrophage recruitment ↓Macrophage phagocytosis ↓TNFα ↓Interleukin 8 ↓Neutrophil recruitment Bloodstream invasion of S pneumoniae Alveolar interstitium Nasopharyngeal cavity ↓Opsonic capacity of IgA ( ) and IgG ( ) Bloodstream Alveolar space Nasopharynx interstitium Figure 1: Alterations in the host response of patient with HIV that might cause increased susceptibility to invasive infection with Streptococcus pneumoniae In the upper airways, impaired recruitment of macrophages due to CD4 T-cell depletion and low concentrations of interleukin 17 enable colonisation by S pneumoniae on the respiratory epithelium. In the lower airways, impaired antipneumococcal activity of IgA and IgG on the mucosal surface of the respiratory epithelium allows the attachment of S pneumoniae to the epithelial cells and migration of the pathogen across the epithelial barrier. Ineffective opsonisation also impairs phagocytosis by alveolar macrophages. Furthermore, alveolar macrophages from HIV-positive patients showed decreased TNFα production in response to TLR2 and TLR4 ligands, and concentrations of TNFα in bronchoalveolar lavage fluid are reduced in patients with HIV, thereby hindering an effective proinflammatory response. Alveolar macrophages from patients with HIV produce lower concentrations of interleukin 8 in response to S pneumoniae, resulting in less effective recruitment of neutrophils. Neutrophils of HIV patients are also less able to respond to chemotactic signals, such as interleukin 8, but also pneumolysin derived from the bacterium, further impairing a massive neutrophil influx. The combination of impairments in antipneumococcal defence enables the invasion of S pneumoniae into the bloodstream. TNFα=tumour necrosis factor α.
  • 5. www.thelancet.com/infection Vol 15 January 2015 99 Review load,74 and positively correlated with CD4 counts.61 Homing of dendritic cells in lymph nodes75 and increased apoptosis76 are possible explanations. Reports on the effect of HIV on dendritic cell maturation are inconsistent. An atypical phenotype of immune activation without full maturation has been reported,56,77 as well as reduced capacity to mature in response to stimuli combined with interleukin 10 induction and immune suppression.78 By contrast, normal maturation in response to stimuli57,73 and HIV- induced maturation have also been described.79 Stimulation-induced production of cytokines, including interleukin 12,57–59 interleukin 6,58 interferon γ,58,59 interferon α,59,61 interleukin 2,60 and interleukin 10,60 was impaired by HIV, as well as dendritic cell- mediated activation of T cells (table 2).79 Reduced interleukin 12 production was associated with impaired activation of NK cells in vitro.59 Some studies, however, reported normal62 or even increased cytokine responses of dendritic cells to stimuli.63 The number of dendritic cells in peripheral blood is reduced in patients with HIV, and, additionally, most studies report impaired dendritic cell function. HIV- induced tolerance to antigen and reduced maturation of dendritic cells might compromise immunity against various of pathogens, rendering patients with HIV more susceptible to sepsis. NK cells and NK T cells Reduced NK cell numbers and function during HIV infection contribute to decreased resistance against HIV and other pathogens (appendix).80,81 Ex-vivo stimulation of NK cells from patients infected with HIV with E coli or Salmonella typhimurium showed that both untreated and treated patients had lower overall frequencies of responsive interferon-γ-producing NK cells (table 2).64 Although this study is the only one to specifically address antibacterial immune responses by NK cells from patients with HIV, many studies report on HIV-induced changes that prevent an effective anti- HIV NK cell response, which might also result in impaired antibacterial defence. First, correlating with viral load, HIV infection induces expansion of a CD56- negative NK cell subpopulation, which is unresponsive to stimulation and is thought to represent an exhausted state, with concomitant loss of responsive CD56- positive NK cell subsets due to increased apoptosis.80,82 CD56-negative NK cells are also defective in their interaction with dendritic cells, thereby contributing to the accumulation of immature dendritic cells.80,83 Second, HIV causes shedding of MHC class I chain- related molecules MICA and MICB from the surface of infected cells. By binding to the NK cell receptor NKG2D, soluble MICA and MICB provide a negative feedback signal, resulting in subsequent down- regulation of NKG2D, thereby promoting the generation of anergic NK cells.82 Third, decreased production of chemokines impairs chemotactic signalling to other immune cells.80,84 NK cells from patients with HIV also showed increased expression of NK-cell inhibitory receptors that prevent lysis of target cells, but these effects were reversed in virally suppressed patients on cART.85,86 No studies on NK T-cell antibacterial defence in HIV-infected hosts were identified, but selective NK T-cell depletion might hinder the proinflammatory response to bacterial pathogens.65 Various mechanisms contribute to expansion of an anergic NK cell population that is less capable of responding to pathogens in individuals with HIV. This expansion might impair direct NK-cell mediated defence against bacteria, and compromise crosstalk with other immune cells. Neutrophils In patients with HIV infection, neutrophils might not be able to function effectively because of several mechanisms (appendix). First, neutropenia is common in patients with HIV.87 Possible mechanisms include increased apoptosis due to the presence of auto- antibodies,88 activation in the absence of secondary infection,89 reduced production attributed to decreased levels of CSF3,90 and treatment-induced neutropenia.49 Additionally, defects in neutrophil function have been described in patients with HIV, including reduced chemotaxis,91,92 impaired transepithelial migration93 (possibly explained by reduced expression of interleukin 8 receptors on neutrophils of these patients92 ), and impaired phagocytosis.40,94–96 In one study96 phagocytosis of S aureus was only impaired when neutrophils were incubated with serum of patients with HIV, indicating reduced opsonisation with antibodies as the mechanism underlying defective phagocytosis. However, in other investigations,94,95 decreased phagocytosis was observed after previous opsonisation in healthy serum, suggesting an antibody- independent mechanism. Although the aforementioned studies used neutrophils from patients with HIV with advanced disease, one study43 recorded an increased capacity for neutrophils to phagocytose E coli and S aureus from patients with early HIV infection. Superoxide production, a mechanism of microbial killing, was increased in unstimulated neutrophils from patients with HIV,43 but the response to pathogens, such as E coli, was reduced.40,89 Additionally, HIV causes impaired C5a (a complement constituent with proinflammatory properties) and interleukin-8-induced degranulation,97 which is associated with reduced expression of C5a and interleukin 8 receptors.92,97 Although HIV causes NET release from neutrophils, it can also counteract this response by inducing production of interleukin 10 by dendritic cells to inhibit NET formation.98 Hence, HIV-induced inhibition of NET formation in response to bacterial pathogens
  • 6. 100 www.thelancet.com/infection Vol 15 January 2015 Review could impair effective host defence. Finally, ex-vivo bacterial killing was reduced,96 especially in patients with advanced disease.96 However, one study94 reported normal killing of S aureus by neutrophils from patients with AIDS. HIV-related defects in neutrophil numbers, chemo- taxis, phagocytosis, superoxide production, cytokine release, bacterial killing, and NET formation have been described, suggesting that HIV renders the host more susceptible to bacterial infection, at least in part by impairing neutrophil-mediated host defence. Cytokines and DAMPs Abundant evidence suggests a change in cytokine profiles in patients with HIV, although very few studies have investigated this change in the context of sepsis (table 2). Two studies50,51 provided comparative data for inflammatory parameters in patients with sepsis admitted to the ICU with or without HIV co-infection. A wide range of cytokines were measured, but no significant differences were noted for most of them. In one study, high concentrations of interleukin 10 were reported in HIV-positive patients compared with HIV- negative patients, which was associated with increased mortality.50 However, the HIV-negative septic controls differed substantially in age and site of infection, which complicates interpretation of these results.50,51 Studies47,99 using plasma from patients with HIV but without sepsis reported increased concentrations of several cytokines implicated in sepsis pathogenesis: interleukin 6, interleukin 10, TNFα, interleukin 1A, interleukin 1β, interleukin 8, CCL2, interleukin 1 receptor antagonist, and, in acute HIV infection, interferon γ. Concentrations of HMGB1 were also increased in patients with HIV.100 However, circulating concentrations of the proinflammatory cytokines interleukin 12, and CSF2, were decreased.49,48 Studies on the effect of cART noted normalisation of most cytokine perturbations.47,48 Raised concentrations of circulating inflammatory markers might in fact be associated with the development of infection; a correlation was identified between raised concentrations of C-reactive protein in patients with HIV and an increased risk of bacterial pneumonia.101 A possible explanation for raised cytokine con- centrations is a primed state of immune cells in patients with HIV causing hyper-responsiveness to stimulation; for example, by microbial products translocated from the gut. Increased TNFα production in response to lipopolysaccharide was observed in PBMCs and monocytes isolated from patients with HIV.38,46 Decreased interleukin 12 concentrations might relate to the type of stimulus and stage of disease, with patients with AIDS showing the most profound deficiency.102 There is abundant evidence for increased circulating concentrations of both proinflammatory and anti-inflammatory cytokines in patients with HIV, combined with ex-vivo evidence for a primed state of immune cells in response to stimulation, which could result in a more profound imbalance between these mediators during sepsis. However, the small number of studies available50,51 in patients with HIV and sepsis reported few differences in cytokine concentrations compared with HIV-negative patients with sepsis. Lymphocytes Overview Antigen-presenting cells interact with lymphocytes to initiate the adaptive immune response. In response to the presentation of antigen, effector CD4 T cells secrete cytokines, such as interferon γ, to enhance phagocytic killing and interaction with B cells to initiate production of antibodies.19 B cells are the cornerstone of immuno- logical memory, and represent the main defence mechanism against reinfection. B cells have also been shown to have an important role in the enhancement of innate immune responses during bacterial sepsis.103 Sepsis causes a substantial depletion of CD4, CD8, and B lymphocytes because of increased apoptosis, which is correlated with sepsis severity.17,104 Because T cells are important in coordinating innate immunity, increased apoptosis of T cells might hinder the proinflammatory response.104 Remaining lymphocytes also show significant reductions in cytokine release on stimulation.105 T lymphocytes During HIV infection, both T-cell numbers and function become compromised (appendix). CD4 T-cell depletion develops through various mechanisms, including direct virus-induced killing, induction of apoptosis, and an inability of the thymus to efficiently compensate for the lost T cells.106 In addition to depletion of CD4 T cells, HIV downregulates the CD4 receptor, which might reduce the immune functions of surviving cells.107 Of particular relevance in the context of sepsis, patients with HIV displayed increased apoptosis of CD3 T cells after in-vitro challenge with S pneumoniae.108 A particular subset of CD4 cells, Th17 cells, which predominate in the gastrointestinal tract and are important in antibacterial defence, are substantially depleted,109 and remaining Th17 cells have an enahanced interleukin 10:TNFα ratio, suggestive of an anti-inflammatory phenotype (table 2).67 Similarly, increased interleukin 10 production was identified in peripheral blood CD4 T cells.66 Mucosal-associated invariant T cells (MAIT cells), a subset of innate-like, tissue-infiltrating lymphocytes that produce interleukin 17, interleukin 22, interferon γ, and TNFα, were also shown to be severely depleted in blood of patients with HIV, and remaining cells showed an activated phenotype combined with functional exhaustion.110,111 Although numbers of MAIT cells in the
  • 7. www.thelancet.com/infection Vol 15 January 2015 101 Review gut were relatively preserved in these studies,110,111 a third study reported MAIT cell depletion in the colon of patients with HIV.112 Since MAIT cells are known to be activated by a wide range of bacteria and fungi,113 their depletion might have an important role in increased susceptibility of patients with HIV to bacterial sepsis. Mucosal depletion of interleukin-17-producing cells, like TH17 cells and MAIT cells, might be related to increased risk of patients with HIV developing non- typhoidal salmonella bacteraemia, because interleukin 17 is known to have an important role in host defence against dissemination of enteric pathogens (figure 2).114 Additionally, interleukin-17-producing CD4 T cells were shown to be essential for recruitment of monocytes and macrophages to allow for effective pneumococcal clearance from the nasopharynx,115 suggesting that CD4 cell depletion has a role in the vulnerability of patients with HIV to the invasive pneumococcal infections (figure 1). Although CD4 T-cell counts decrease, CD8 T-cell numbers expand during HIV infection.116 Nonetheless, both T-cell compartments display a phenotype of immune activation and exhaustion, as suggested by heightened expression of the activation markers HLA- DR and CD38, and of the inhibitory receptors PDCD1 and CTLA4 in untreated patients with HIV.117,118 Furthermore, the CD8 T-cell compartment has been shown to arrest at a late differentiated phenotype,119 and is unable to mount an effective cytolytic response.120,121 Both reduction in CD4 T-cell numbers, and functional impairments of CD4 T cells and CD8 T cells might contribute to increased susceptibility of patients with HIV to develop invasive bacterial infections and sepsis. Most of these defects can be restored by cART,106 but impaired restoration of the CD4 T-cell proliferation response has been described in patients with HIV with lower nadir CD4 T-cell counts before therapy,122 thus providing one possible explanation for higher rates of bacteraemia in cART-treated patients compared with HIV-negative people. B-lymphocytes In patients with HIV, several defects in B-cell function have been detected, which could have a role in increased susceptibility to infections, especially in patients with non-typhoidal salmonella and S pneumoniae (appendix). Functional disturbances include hypergammaglo- bulinaemia, polyclonal activation and exhaustion, impaired class-switch recombination, dysfunctional interaction between T cells and B cells, blunted proliferation response to both T-cell-dependent and T-cell-independent antigens, poor immune responses against vaccination antigens, short duration of antibody response induced by pneumococcal vaccination, and increased apoptosis.123–127 Additionally, patients with HIV show changes in the distribution of B-cell subsets. Resting memory B cells are severely depleted, whereas transitional B cells are expanded.123 An aberrant B-cell population, CD21low CD27− B cells, normally present in very low numbers in peripheral blood, was reported to rise in the blood of viraemic patients with HIV proportional to viral load,128 which showed features of immune activation and cellular exhaustion.129 Although changes in most B-cell subtypes are restored by cART, resting memory B cells usually remain depleted128 because they can only be preserved when cART is started early in infection.130 Additionally, IgG concentrations were shown to remain raised despite long-term cART in 45% of patients, indicating continuous B-cell activation.131 B-cell dysfunction probably has an important role in the increased risk of patients with HIV developing invasive infections with S pneumoniae (figure 1) and non-typhoidal salmonella (figure 2). Patients with HIV have reduced numbers of pneumococcal antigen- specific IgG antibody-secreting cells and lower concentrations of pneumococcal antibodies.124,132 Additionally, IgA in the epithelial lining fluid of the lung showed impaired antipneumococcal activity.133 Although some studies reported normal,134 or even Th17 ↓Interleukin 17 Intestinal lumen Lamina propria Bloodstream Interleukin 17 NTS in the gut of a host without HIV infection NTS in the gut of an HIV-infected host Th17 depletion and reduced interleukin 17 compromise gut barrier function Inhibitory antibodies bind to LPS, which prevents bacterial killing Binding of IgG to outer membrane proteins enables bacterial lysis and killing C1q LPS MAC Figure2: HIV-induced changes in host defence against non-typhoidal salmonella (NTS) infection fromthe gut Normally, interleukin-17-producing Th17 cells effectively prevent invasion of NTS from the gut (left side), but HIV infection results in Th17 cell depletion and reduced interleukin 17 concentrations, thereby compromising the gut barrier function against NTS (right side). If NTS bacteria manage to reach the bloodstream in a host without HIV infection, they are effectively opsonised by IgG antibodies that bind to the outer membrane protein of NTS and enable complement (C1q) binding, followed by formation of a MAC on the bacterial surface, resulting in lysis and killing of the bacteria (left side). By contrast, in the patient with HIV, NTS bacteria are opsonised by antibodies that have an inhibitory effect on bacterial killing. They do not bind to the outer membrane protein of the NTS, but to its LPS, which does not enable lysis and killing, and allows for survival of NTS in the bloodstream (right side). NTS=non-typhoid salmonella. MAC=membrane attack complex. LPS=lipopolysaccharide.
  • 8. 102 www.thelancet.com/infection Vol 15 January 2015 Review increased concentrations of IgG anti-pneumococcal- specific antibodies in patients with HIV,135 the opsonic capacity of these antibodies was reduced.135 In the case of NTS, IgG hypersecretion might be implicated in the increased risk of infection in patients with HIV; serum from African patients with HIV contained high concentrations of IgG antibodies against non-typhoidal salmonella that prevented destruction of Salmonella spp.136 These antibodies had non-neutralising activity against non-typhoidal salmonella lipopolysaccharide and exerted a dose-dependent inhibitory effect on the killing capacity of healthy serum, suggesting an important role for anti-salmonella LPS IgG in the prevention of clearance of non-typhoidal salmonella in patients with HIV (figure 2).137 Perturbations in humoral immunity probably have a substantial role in the increased susceptibility of patients with HIV to bacterial infections, with simultaneous depletion of the memory B-cell com- partment, and inadequate activation, IgG production, and B-cell exhaustion, which are only partly restored by cART. The coagulation system Patients with sepsis invariably show a disturbed balance between coagulation and anticoagulation.16,19 In severe cases, this disturbance results in disseminated intra- vascular coagulation, while at the same time, a consumptive coagulopathy develops, causing increased risk of bleeding. disseminated intravascular coagulation is triggered by a fulminant host response to infection, causing aberrant expression of tissue factor, impairment of physiological anticoagulant pathways due to endothelial dysfunction, and the suppression of fibrinolysis due to overproduction of SERPINE1 by endothelial cells.16 Several abnormalities in the coagulation system and endothelial cell function of patients with HIV have been identified (figure 3). In general, HIV-induced alterations in haemostasis are remarkably similar to those described in sepsis. First, patients with HIV display a procoagulant state, as shown by their increased risk of thromboembolic events.137 Coagulation activation in these patients is further suggested by increased concentrations of D-dimer and fibrinogen,138,139 increased platelet activation,140 and increased expression of tissue factor on platelets and monocytes.141,142 Anticoagulation is hindered by reduced concentrations of protein C, and protein S,138,143 and fibrinolysis might be impaired as a consequence of increased SERPINE concentrations in patients with HIV.139,143 Last, ample evidence shows endothelial activation and dysfunction in patients with HIV. Raised concentrations of VWF, soluble ICAM1, and soluble VCAM1 have been reported in several studies.138,143,144 Additionally, endothelial function, assessed by measuring flow-mediated dilation of the brachial artery, was impaired in patients with HIV.145 Autopsy studies of patients with HIV, with or without AIDS-related complications, also reported endothelial injury.146,147 Finally, a study on angiogenic factors in severe bacterial infection in Malawian children noted that HIV-positive cases had significantly higher concentrations of ANGPT2 compared with HIV- negative children with similar illness.148 High con- centrations of ANGPT2, an angiogenic peptide that increases endothelial activation and vascular permeability, are associated with disseminated intra- vascular coagulation and mortality in sepsis.148 Studies on the effect of cART are inconsistent, with some recording complete normalisation144 and others reporting partial improvement,136,149 as well as studies describing a detrimental effect of cART, especially regimens containing protease inhibitors.137,150 At present, no studies on the effect of HIV infection on the procoagulant response to sepsis exist. In view of the disturbed haemostatic balance in patients with HIV, with alterations that strongly resemble those detected in sepsis, it is conceivable that HIV causes an even greater disruption of procoagulant and anticoagulant mechanisms during sepsis. The complement system The complement system is an important part of the first-line host defence against bacteria, by opsonisation, lysis of pathogens and infected cells, and production of chemoattractants. Clinical and experimental sepsis are ↑ANGPT2 ↑sICAM1 ↑sVCAM1 ↑VWF ↑Tissue factor Coagulation cascade ↑Fibrinogen Fibrin Thrombus formation↑D-dimer Activated protein C ↓Protein C ↓Protein S ↑SERPINE ↓Fibrinolysis Increased coagulation Impaired anticoagulation and fibrinolysis Endothelial activation and damage Figure3:HIV-inducedchangesinthecoagulationsystemandendotheliumresemblesepsis-inducedcoagulopathy Expression of tissue factor on circulating monocytes is increased in patients with HIV, which triggers the extrinsic pathway of the coagulation cascade, eventually resulting in the transformation of fibrinogen to fibrin. Increased concentrations of fibrinogen and D-dimer, a fibrin degradation product, are seen in patients with HIV, providing further evidence for coagulation activation. The coagulation cascade is normally balanced by the anticoagulant system, which includes activated protein C. However, in patients with HIV protein C, and protein S, which is needed to activate protein C, are depleted. Additionally, fibrinolysis is inhibited by increased concentrations of SERPINE, further facilitating thrombus formation in patient with HIV. The endothelium of patients with HIV becomes activated and damaged. Secretion of sICAM1, sVCAM1, and VWF are increased, and the endothelium shows an irregular pattern with multinucleated endothelial cells and increased monocyte adhesion. Higher concentrations of ANGPT2 are also seen in patients with HIV,and are known to cause endothelial cell death and disrupt the endothelial monolayer. Notably, all these changes are very similar to those described in patients with sepsis. sICAM-1=soluble ICAM1. sVCAM1=soluble VCAM1.
  • 9. www.thelancet.com/infection Vol 15 January 2015 103 Review associated with activation of the complement system, as shown by increased plasma concentrations of com- plement constituents C3a, C4a, and C5a.21 Although complement is essential in the host defence against bacteria, increased circulating C3a and C5a are likely to contribute to sepsis-induced tissue damage, multiorgan failure, and septic shock, because of their strong proinflammatory functions.151 C5a is important for the outcome of experimental sepsis, as reported by studies in which treatment with an anti-C5a antibody improved haemodynamic parameters, attenuated coagulopathy, and improved organ function.21 Although the complement system is generally known for its activity against bacterial pathogens, many reports describe HIV-induced activation of the complement system through the the classic pathway via interaction of gp41 with C1q;152 interaction of HIV with mannose- binding lectin, the triggering molecule of the lectin pathway;152 and activation of the alternative pathway through C3 binding of HIV-infected monocytes and lymphocytes.153 After seroconversion, HIV-specific antibodies further enhance complement activation via the classic pathway,152 and raised concentrations of circulating C5a have been recorded in patients with HIV.91 However, HIV can escape complement-induced lysis by acquiring regulators of complement activation, which allow the virus to use the complement system for transport to the lymphoid system and infection of susceptible cells.152 Increased complement activation might contribute to the generalised immune activation observed in HIV and, hypothetically, have a detrimental role during sepsis pathogenesis by disturbing the balance between proinflammatory and anti-inflammatory mechanisms. However, at present, studies investigating the effect of HIV infection on complement activation in sepsis have not been reported. Future perspectives The present treatment of sepsis is based on antibiotics and supportive care.154 In past decades, many clinical trials have been done to investigate inhibition of the abundant inflammatory response generally held responsible for sepsis mortality.19,21 In more recent years, however, attention has shifted to findings that patients with sepsis invariably show evidence for immune suppression, which has been implicated as an important cause of secondary infections and late mortality.17 Preclinical studies have suggested that immune stimulatory therapy, which aims to overcome sepsis-induced immune suppression, could improve sepsis outcome. Interventions assessed in this context include immunomodulatory cytokines, such as interleukin 7, interleukin 15, and CSF2, as well as antibodies targeting co-inhibitory molecules, like PDCD1, CD274, and CTLA417,155 (table 3).156,157 Notably, similar strategies have been suggested to remedy Relevance in sepsis Relevance in HIV Interleukin 7 Prevented apoptosis-inducedT-cell depletion, and reversed sepsis-induced depressionofT-cell cytokines in a murine sepsis model Improved lymphocyte function after ex-vivo treatmentof cells from septic patients Increase in functional naive and central memoryCD4 andCD8T cells in clinicaltrials Interleukin 15 Induces proliferation and activationof T cells, natural killer cells and natural killer T cells Decreased apoptosis and improved survival in a murine sepsis model Theoretically,the same beneficial effects as in sepsis might beobserved; however,delayed viral suppression and failureto reconstitute CD4T cells in SIV-infected macaquestreated with interleukin 15discouraged further research into interleukin 15 as an immunomodulatorydrug CSF2 Intargeted patient groups (thosewith low HLA-DR expression inone study, and low ex-vivotumour necrosis factor α producing capacity)CSF2 increasedthe functionof innate immune cells and improved clinical outcomes CSF2duringART interruptions blunted viral rebound and preventedthedecreaseofCD4 cell counts156 PDCD1/CD274 Blockageof PDCD1or its ligand improved survival in murine sepsis studies Blockageof PDCD1 causedCD8T-cell and B-cell activation, reduced viral load, and improved survival in an SIV-macaque model. PDCD1 blockade reduced hyperimmune activation and microbialtranslocation in a SIV-macaque model CD274 blockade causedCD4T-cell restoration in an ex-vivo studyusing PBMCs from patients with HIV157 CTLA4 Anti-CTLA4treatment improved sepsis- induced lymphocyte apoptosis and survival from secondary fungal infections in a murine sepsis model Anti-CTLA4 treatment caused an increase in CD4 and CD8 T cell responses and reduced viral RNA in a SIV-macaque model However, a second study with a similar design reported no expansion of SIV specific T cells and increased viral replication at mucosal sites Information in this table was derived from Hotchkiss et al and Hutchins et al17,155 for sepsis, andVanham andVan Gulck for HIV,158 unless otherwise indicated in the table. ART=antiretroviral therapy. PBMC=peripheral blood mononuclear cell. SIV=simian immunodeficiency virus. Table 3: Parallels between the use of immunomodulatory agents in sepsis and HIV Search strategy and selection criteria We searched PubMed using the following search string: (“HIV” [MeSH Terms] or “acquired immunodeficiency syndrome” [MeSH Terms]) and (“immunity” [MeSH Terms] or “monocytes” [MeSH Terms] or “macrophages” [MeSH Terms] or “dendritic cell” [MeSH Terms] or “neutrophils” [MeSH Terms] or “pattern recognition receptors” [MeSH Terms] or “lymphocytes” [MeSH Terms] or “cytokines” [MeSH Terms] or “complement system proteins” [MeSH Terms] or “blood coagulation” [MeSH Terms] or “disseminated intravascular coagulation” [MeSH Terms]) and (“bacteria” [MeSH Terms] or “sepsis” [MesH Terms] or “bacteremia” [Mesh Terms)”. We also combined the MeSH Terms for “salmonella” and “S pneumoniae” with the MeSH Terms for “HIV” or “AIDS”. The search was limited to English-language articles published between Jan 1, 1990 and June 1, 2014. We reviewed relevant articles identified in this search, papers cited in these studies, and articles from the authors’ personal files.
  • 10. 104 www.thelancet.com/infection Vol 15 January 2015 Review HIV-induced immune suppression (table 3).158 Pro- inflammatory and procoagulant pathways targeted by novel sepsis therapies under clinical evaluation, such as anti-HMGB1 strategies and soluble thrombo- modulin,159,160 could be of interest for patients with HIV and sepsis because HIV by itself already adversely affects these mechanisms. Many pathogenic mecha- nisms between sepsis and HIV overlap, so it is conceivable that patients with HIV presenting with sepsis form a group that will particularly benefit from new drugs targeting septic immunosuppression. In this light, the systematic exclusion of patients with HIV from sepsis trials might need to be reconsidered. Conclusion In the era of effective cART, bacterial sepsis has evolved as a major cause of mortality in patients with HIV. HIV infection affects many components of the immune response, which on the one hand renders the HIV- infected host more susceptible to invasive infection, and on the other hand might further exacerbate the deregulated host response to sepsis. Specific research on sepsis in patients with HIV is scarce and future studies are needed to gain more insight into the particularities of sepsis pathogenesis in these patients. Although cART greatly improves patients’ immune function, as shown by the strong reduction in opportunistic infections, patients continue to have an increased risk of developing invasive bacterial infection, suggesting the current and future relevance of this under-researched area. Declaration of interests We declare no competing interests. Contributors MAH conceived the idea, searched the scientific literature, interpreted the data, wrote the manuscript and created the figures. MPG and TvdP. interpreted the data and wrote the manuscript. References 1 Kim JH, Psevdos G Jr, Gonzalez E, Singh S, Kilayko MC, Sharp V. All-cause mortality in hospitalized HIV-infected patients at an acute tertiary care hospital with a comprehensive outpatient HIV care program in New York City in the era of highly active antiretroviral therapy (HAART). Infection 2013; 41: 545–51. 2 Gilks CF, Ojoo SA, Ojoo JC, et al. Invasive pneumococcal disease in a cohort of predominantly HIV-1 infected female sex-workers in Nairobi, Kenya. Lancet 1996; 347: 718–23. 3 Huson MA, Stolp SM, van der Poll T, Grobusch MP. Community- acquired bacterial bloodstream infections in HIV-infected patients: a systematic review. Clin Infect Dis 2014; 58: 79–92. 4 Jordano Q, Falcó V, Almirante B, et al. Invasive pneumococcal disease in patients infected with HIV: still a threat in the era of highly active antiretroviral therapy. Clin Infect Dis 2004; 38: 1623–28. 5 Archibald LK, den Dulk MO, Pallangyo KJ, Reller LB. Fatal Mycobacterium tuberculosis bloodstream infections in febrile hospitalized adults in Dar es Salaam, Tanzania. Clin Infect Dis 1998; 26: 290–96. 6 Archibald LK, McDonald LC, Nwanyanwu O, et al. A hospital- based prevalence survey of bloodstream infections in febrile patients in Malawi: implications for diagnosis and therapy. J Infect Dis 2000; 181: 1414–20. 7 Arthur G, Nduba VN, Kariuki SM, Kimari J, Bhatt SM, Gilks CF. Trends in bloodstream infections among human immunodeficiency virus-infected adults admitted to a hospital in Nairobi, Kenya, during the last decade. Clin Infect Dis 2001; 33: 248–56. 8 Bell M, Archibald LK, Nwanyanwu O, et al. Seasonal variation in the etiology of bloodstream infections in a febrile inpatient population in a developing country. Int J Infect Dis 2001; 5: 63–69. 9 Crump JA, Ramadhani HO, Morrissey AB, et al. Invasive bacterial and fungal infections among hospitalized HIV-infected and HIV- uninfected adults and adolescents in northern Tanzania. Clin Infect Dis 2011; 52: 341–48. 10 Grant AD, Djomand G, Smets P, et al. Profound immunosuppression across the spectrum of opportunistic disease among hospitalized HIV-infected adults in Abidjan, Côte d’Ivoire. AIDS 1997; 11: 1357–64. 11 Mayanja BN, Todd J, Hughes P, et al. Septicaemia in a population- based HIV clinical cohort in rural Uganda, 1996–2007: incidence, aetiology, antimicrobial drug resistance and impact of antiretroviral therapy. Trop Med Int Health 2010; 15: 697–705. 12 Meremo A, Mshana SE, Kidenya BR, Kabangila R, Peck R, Kataraihya JB. High prevalence of Non-typhoid salmonella bacteraemia among febrile HIV adult patients admitted at a tertiary Hospital, North-Western Tanzania. Int Arch Med 2012; 5: 28. 13 Nadjm B, Mtove G, Amos B, et al. Severe febrile illness in adult hospital admissions in Tanzania: a prospective study in an area of high malaria transmission. Trans R Soc Trop Med Hyg 2012; 106: 688–95. 14 Peters RP, Zijlstra EE, Schijffelen MJ, et al. A prospective study of bloodstream infections as cause of fever in Malawi: clinical predictors and implications for management. Trop Med Int Health 2004; 9: 928–34. 15 Ssali FN, Kamya MR, Wabwire-Mangen F, et al. A prospective study of community-acquired bloodstream infections among febrile adults admitted to Mulago Hospital in Kampala, Uganda. J Acquir Immune Defic Syndr Hum Retrovirol 1998; 19: 484–89. 16 Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med 2013; 369: 840–51. 17 Hotchkiss RS, Monneret G, Payen D. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis 2013; 13: 260–68. 18 Appay V, Sauce D. Immune activation and inflammation in HIV-1 infection: causes and consequences. J Pathol 2008; 214: 231–41. 19 Stearns-Kurosawa DJ, Osuchowski MF, Valentine C, Kurosawa S, Remick DG. The pathogenesis of sepsis. Annu Rev Pathol 2011; 6: 19–48. 20 Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell 2006; 124: 783–801. 21 van der Poll T, Opal SM. Host-pathogen interactions in sepsis. Lancet Infect Dis 2008; 8: 32–43. 22 Mancuso G, Gambuzza M, Midiri A, et al. Bacterial recognition by TLR7 in the lysosomes of conventional dendritic cells. Nat Immunol 2009; 10: 587–94. 23 Cavassani KA, Ishii M, Wen H, et al. TLR3 is an endogenous sensor of tissue necrosis during acute inflammatory events. J Exp Med 2008; 205: 2609–21. 24 Murphy KM. Janeway’s immunobiology, 8th edn. New York: Garland Science; 2012. 25 Chiche L, Forel JM, Thomas G, et al. The role of natural killer cells in sepsis. J Biomed Biotechnol 2011: 986491. 26 Souza-Fonseca-Guimaraes F, Adib-Conquy M, Cavaillon JM. Natural killer (NK) cells in antibacterial innate immunity: angels or devils? Mol Med 2012; 18: 270–85. 27 Leung B, Harris HW. NKT cells: the culprits of sepsis? J Surg Res 2011; 167: 87–95. 28 Nathan C. Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol 2006; 6: 173–82. 29 de Jong HK, van der Poll T, Wiersinga WJ. The systemic pro- inflammatory response in sepsis. J Innate Immun 2010; 2: 422–30. 30 Yang H, Antoine DJ, Andersson U, Tracey KJ. The many faces of HMGB1: molecular structure-functional activity in inflammation, apoptosis, and chemotaxis. J Leukoc Biol 2013; 93: 865–73. 31 Epple HJ, Zeitz M. HIV infection and the intestinal mucosal barrier. Ann N Y Acad Sci 2012; 1258: 19–24. 32 Brenchley JM, Price DA, Schacker TW, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med 2006; 12: 1365–71.
  • 11. www.thelancet.com/infection Vol 15 January 2015 105 Review 33 Sanders CM, Cruse JM, Lewis RE. Toll-like receptors, cytokines and HIV-1. Exp Mol Pathol 2008; 84: 31–36. 34 Scagnolari C, Selvaggi C, Chiavuzzo L, et al. Expression levels of TLRs involved in viral recognition in PBMCs from HIV-1-infected patients failing antiretroviral therapy. Intervirology 2009; 52: 107–14. 35 Hernández JC, Stevenson M, Latz E, Urcuqui-Inchima S. HIV type 1 infection up-regulates TLR2 and TLR4 expression and function in vivo and in vitro. AIDS Res Hum Retroviruses 2012; 28: 1313–28. 36 Heggelund L, Müller F, Lien E, et al. Increased expression of toll- like receptor 2 on monocytes in HIV infection: possible roles in inflammation and viral replication. Clin Infect Dis 2004; 39: 264–69. 37 Gordon SB, Jagoe RT, Jarman ER, et al. The alveolar microenvironment of patients infected with human immunodeficiency virus does not modify alveolar macrophage interactions with Streptococcus pneumoniae. Clin Vaccine Immunol 2013; 20: 882–91. 38 Lester RT, Yao XD, Ball TB, et al. Toll-like receptor expression and responsiveness are increased in viraemic HIV-1 infection. AIDS 2008; 22: 685–94. 39 Schuitemaker H, Kootstra NA, Fouchier RA, Hooibrink B, Miedema F. Productive HIV-1 infection of macrophages restricted to the cell fraction with proliferative capacity. EMBO J 1994; 13: 5929–36. 40 Michailidis C, Giannopoulos G, Vigklis V, Armenis K, Tsakris A, Gargalianos P. Impaired phagocytosis among patients infected by the human immunodeficiency virus: implication for a role of highly active anti-retroviral therapy. Clin Exp Immunol 2012; 167: 499–504. 41 Trial J, Birdsall HH, Hallum JA, et al. Phenotypic and functional changes in peripheral blood monocytes during progression of human immunodeficiency virus infection. Effects of soluble immune complexes, cytokines, subcellular particulates from apoptotic cells, and HIV-1-encoded proteins on monocytes phagocytic function, oxidative burst, transendothelial migration, and cell surface phenotype. J Clin Invest 1995; 95: 1690–701. 42 Nottet HS, de Graaf L, de Vos NM, et al. Phagocytic function of monocyte-derived macrophages is not affected by human immunodeficiency virus type 1 infection. J Infect Dis 1993; 168: 84–91. 43 Bandres JC, Trial J, Musher DM, Rossen RD. Increased phagocytosis and generation of reactive oxygen products by neutrophils and monocytes of men with stage 1 human immunodeficiency virus infection. J Infect Dis 1993; 168: 75–83. 44 Brown JN, Kohler JJ, Coberley CR, Sleasman JW, Goodenow MM. HIV-1 activates macrophages independent of Toll-like receptors. PLoS One 2008; 3: e3664. 45 Gekonge B, Giri MS, Kossenkov AV, et al. Constitutive gene expression in monocytes from chronic HIV-1 infection overlaps with acute Toll-like receptor induced monocyte activation profiles. PLoS One 2012; 7: e41153. 46 Dutertre CA, Amraoui S, DeRosa A, et al. Pivotal role of M-DC8+ monocytes from viremic HIV-infected patients in TNFα overproduction in response to microbial products. Blood 2012; 120: 2259–68. 47 Haissman JM, Vestergaard LS, Sembuche S, et al. Plasma cytokine levels in Tanzanian HIV-1-infected adults and the effect of antiretroviral treatment. J Acquir Immune Defic Syndr 2009; 52: 493–97. 48 Keating SM, Golub ET, Nowicki M, et al, and the Women’s Interagency HIV Study. The effect of HIV infection and HAART on inflammatory biomarkers in a population-based cohort of women. AIDS 2011; 25: 1823–32. 49 Kuritzkes DR. Neutropenia, neutrophil dysfunction, and bacterial infection in patients with human immunodeficiency virus disease: the role of granulocyte colony-stimulating factor. Clin Infect Dis 2000; 30: 256–60. 50 Silva JM Jr, dos Santos SS. Sepsis in AIDS patients: clinical, etiological and inflammatory characteristics. J Int AIDS Soc 2013; 16: 17344. 51 Amancio RT, Japiassu AM, Gomes RN, et al. The innate immune response in HIV/AIDS septic shock patients: a comparative study. PLoS One 2013; 8: e68730. 52 Lester RT, Yao XD, Ball TB, et al. HIV-1 RNA dysregulates the natural TLR response to subclinical endotoxemia in Kenyan female sex-workers. PLoS One 2009; 4: e5644. 53 Tachado SD, Zhang J, Zhu J, Patel N, Koziel H. HIV impairs TNF-alpha release in response to Toll-like receptor 4 stimulation in human macrophages in vitro. Am J Respir Cell Mol Biol 2005; 33: 610–21. 54 Nicol MQ, Mathys JM, Pereira A, Ollington K, Ieong MH, Skolnik PR. Human immunodeficiency virus infection alters tumor necrosis factor alpha production via Toll-like receptor- dependent pathways in alveolar macrophages and U1 cells. J Virol 2008; 82: 7790–98. 55 Gordon SB, Jarman ER, Kanyanda S, et al. Reduced interleukin-8 response to StreptococcuS pneumoniae by alveolar macrophages from adults with HIV/AIDS. AIDS 2005; 19: 1197–200. 56 Denis M, Ghadirian E. Dysregulation of interleukin 8, interleukin 10, and interleukin 12 release by alveolar macrophages from HIV type 1-infected subjects. AIDS Res Hum Retroviruses 1994; 10: 1619–27. 57 Smed-Sörensen A, Loré K, Walther-Jallow L, Andersson J, Spetz AL. HIV-1-infected dendritic cells up-regulate cell surface markers but fail to produce IL-12 p70 in response to CD40 ligand stimulation. Blood 2004; 104: 2810–17. 58 Frleta D, Ochoa CE, Kramer HB, et al. HIV-1 infection-induced apoptotic microparticles inhibit human DCs via CD44. J Clin Invest 2012; 122: 4685–97. 59 Benlahrech A, Gotch F, Kelleher P, Patterson S. Loss of NK stimulatory capacity by plasmacytoid and monocyte-derived DC but not myeloid DC in HIV-1 infected patients. PLoS One 2011; 6: e17525. 60 Donaghy H, Gazzard B, Gotch F, Patterson S. Dysfunction and infection of freshly isolated blood myeloid and plasmacytoid dendritic cells in patients infected with HIV-1. Blood 2003; 101: 4505–11. 61 Feldman S, Stein D, Amrute S, et al. Decreased interferon-alpha production in HIV-infected patients correlates with numerical and functional deficiencies in circulating type 2 dendritic cell precursors. Clin Immunol 2001; 101: 201–10. 62 Chougnet C, Cohen SS, Kawamura T, et al. Normal immune function of monocyte-derived dendritic cells from HIV-infected individuals: implications for immunotherapy. J Immunol 1999; 163: 1666–73. 63 Sabado RL, O’Brien M, Subedi A, et al. Evidence of dysregulation of dendritic cells in primary HIV infection. Blood 2010; 116: 3839–52. 64 Dillon SM, Lee EJ, Bramante JM, Barker E, Wilson CC. The natural killer cell interferon-gamma response to bacteria is diminished in untreated HIV-1 infection and defects persist despite viral suppression. J Acquir Immune Defic Syndr 2014; 65: 259–67. 65 Vasan S, Tsuji M. A double-edged sword: the role of NKT cells in malaria and HIV infection and immunity. Semin Immunol 2010; 22: 87–96. 66 Ostrowski MA, Gu JX, Kovacs C, Freedman J, Luscher MA, MacDonald KS. Quantitative and qualitative assessment of human immunodeficiency virus type 1 (HIV-1)-specific CD4+ T cell immunity to gag in HIV-1-infected individuals with differential disease progression: reciprocal interferon-gamma and interleukin-10 responses. J Infect Dis 2001; 184: 1268–78. 67 Kim CJ, McKinnon LR, Kovacs C, et al. Mucosal Th17 cell function is altered during HIV infection and is an independent predictor of systemic immune activation. J Immunol 2013; 191: 2164–73. 68 Jambo KC, Banda DH, Kankwatira AM, et al. Small alveolar macrophages are infected preferentially by HIV and exhibit impaired phagocytic function. Mucosal Immunol 2014; 7: 1116–26. 69 van der Poll T, Keogh CV, Buurman WA, Lowry SF. Passive immunization against tumor necrosis factor-alpha impairs host defense during pneumococcal pneumonia in mice. Am J Respir Crit Care Med 1997; 155: 603–08. 70 Olliver M, Hiew J, Mellroth P, Henriques-Normark B, Bergman P. Human monocytes promote Th1 and Th17 responses to Streptococcus pneumoniae. Infect Immun 2011; 79: 4210–17.
  • 12. 106 www.thelancet.com/infection Vol 15 January 2015 Review 71 Dockrell DH, Marriott HM, Prince LR, et al. Alveolar macrophage apoptosis contributes to pneumococcal clearance in a resolving model of pulmonary infection. J Immunol 2003; 171: 5380–88. 72 Patel NR, Zhu J, Tachado SD, et al. HIV impairs TNF-alpha mediated macrophage apoptotic response to Mycobacterium tuberculosis. J Immunol 2007; 179: 6973–80. 73 Dillon SM, Robertson KB, Pan SC, et al. Plasmacytoid and myeloid dendritic cells with a partial activation phenotype accumulate in lymphoid tissue during asymptomatic chronic HIV-1 infection. J Acquir Immune Defic Syndr 2008; 48: 1–12. 74 Donaghy H, Pozniak A, Gazzard B, et al. Loss of blood CD11c(+) myeloid and CD11c(-) plasmacytoid dendritic cells in patients with HIV-1 infection correlates with HIV-1 RNA virus load. Blood 2001; 98: 2574–76. 75 Loré K, Sönnerborg A, Broström C, et al. Accumulation of DC-SIGN+CD40+ dendritic cells with reduced CD80 and CD86 expression in lymphoid tissue during acute HIV-1 infection. AIDS 2002; 16: 683–92. 76 Dillon SM, Friedlander LJ, Rogers LM, et al. Blood myeloid dendritic cells from HIV-1-infected individuals display a proapoptotic profile characterized by decreased Bcl-2 levels and by caspase-3+ frequencies that are associated with levels of plasma viremia and T cell activation in an exploratory study. J Virol 2011; 85: 397–409. 77 Izmailova E, Bertley FM, Huang Q, et al. HIV-1 Tat reprograms immature dendritic cells to express chemoattractants for activated T cells and macrophages. Nat Med 2003; 9: 191–97. 78 Granelli-Piperno A, Golebiowska A, Trumpfheller C, Siegal FP, Steinman RM. HIV-1-infected monocyte-derived dendritic cells do not undergo maturation but can elicit IL-10 production and T cell regulation. Proc Natl Acad Sci USA 2004; 101: 7669–74. 79 Yonkers NL, Rodriguez B, Asaad R, Lederman MM, Anthony DD. Systemic immune activation in HIV infection is associated with decreased MDC responsiveness to TLR ligand and inability to activate naive CD4 T-cells. PLoS One 2011; 6: e23884. 80 Funke J, Dürr R, Dietrich U, Koch J. Natural killer cells in HIV-1 infection: a double-edged sword. AIDS Rev 2011; 13: 67–76. 81 Azzoni L, Papasavvas E, Chehimi J, et al. Sustained impairment of IFN-gamma secretion in suppressed HIV-infected patients despite mature NK cell recovery: evidence for a defective reconstitution of innate immunity. J Immunol 2002; 168: 5764–70. 82 Jost S, Altfeld M. Evasion from NK cell-mediated immune responses by HIV-1. Microbes Infect 2012; 14: 904–15. 83 Mavilio D, Lombardo G, Kinter A, et al. Characterization of the defective interaction between a subset of natural killer cells and dendritic cells in HIV-1 infection. J Exp Med 2006; 203: 2339–50. 84 Kottilil S, Chun TW, Moir S, et al. Innate immunity in human immunodeficiency virus infection: effect of viremia on natural killer cell function. J Infect Dis 2003; 187: 1038–45. 85 Kottilil S, Shin K, Planta M, et al. Expression of chemokine and inhibitory receptors on natural killer cells: effect of immune activation and HIV viremia. J Infect Dis 2004; 189: 1193–98. 86 Mavilio D, Benjamin J, Daucher M, et al. Natural killer cells in HIV-1 infection: dichotomous effects of viremia on inhibitory and activating receptors and their functional correlates. Proc Natl Acad Sci USA 2003; 100: 15011–16. 87 Munyazesa E, Emile I, Mutimura E, et al. Assessment of haematological parameters in HIV-infected and uninfected Rwandan women: a cross-sectional study. BMJ Open 2012; 2: e001600. 88 Rubinstein DB, Farrington GK, O’Donnell C, Hartman KR, Wright DG. Autoantibodies to leukocyte alphaMbeta2 integrin glycoproteins in HIV infection. Clin Immunol 1999; 90: 352–59. 89 Elbim C, Prevot MH, Bouscarat F, et al. Polymorphonuclear neutrophils from human immunodeficiency virus-infected patients show enhanced activation, diminished fMLP-induced L-selectin shedding, and an impaired oxidative burst after cytokine priming. Blood 1994; 84: 2759–66. 90 Mauss S, Steinmetz HT, Willers R, et al. Induction of granulocyte colony-stimulating factor by acute febrile infection but not by neutropenia in HIV-seropositive individuals. J Acquir Immune Defic Syndr Hum Retrovirol 1997; 14: 430–34. 91 Meddows-Taylor S, Pendle S, Tiemessen CT. Altered expression of CD88 and associated impairment of complement 5a-induced neutrophil responses in human immunodeficiency virus type 1-infected patients with and without pulmonary tuberculosis. J Infect Dis 2001; 183: 662–65. 92 Meddows-Taylor S, Martin DJ, Tiemessen CT. Reduced expression of interleukin-8 receptors A and B on polymorphonuclear neutrophils from persons with human immunodeficiency virus type 1 disease and pulmonary tuberculosis. J Infect Dis 1998; 177: 921–30. 93 Hofman P, Fischer F, Far DF, et al. Impairment of HIV polymorphonuclear leukocyte transmigration across T84 cell monolayers: an alternative mechanisms for increased intestinal bacterial infections in AIDS? Eur Cytokine Netw 1999; 10: 373–82. 94 Pos O, Stevenhagen A, Meenhorst PL, Kroon FP, Van Furth R. Impaired phagocytosis of StaphylococcuS aureus by granulocytes and monocytes of AIDS patients. Clin Exp Immunol 1992; 88: 23–28. 95 Schaumann R, Krosing J, Shah PM. Phagocytosis of Escherichia coli and StaphylococcuS aureus by neutrophils of human immunodeficiency virus-infected patients. Eur J Med Res 1998; 3: 546–48. 96 Roilides E, Mertins S, Eddy J, Walsh TJ, Pizzo PA, Rubin M. Impairment of neutrophil chemotactic and bactericidal function in children infected with human immunodeficiency virus type 1 and partial reversal after in vitro exposure to granulocyte- macrophage colony-stimulating factor. J Pediatr 1990; 117: 531–40. 97 Meddows-Taylor S, Kuhn L, Meyers TM, Sherman G, Tiemessen CT. Defective neutrophil degranulation induced by interleukin-8 and complement 5a and down-regulation of associated receptors in children vertically infected with human immunodeficiency virus type 1. Clin Diagn Lab Immunol 2001; 8: 21–30. 98 Saitoh T, Komano J, Saitoh Y, et al. Neutrophil extracellular traps mediate a host defense response to human immunodeficiency virus-1. Cell Host Microbe 2012; 12: 109–16. 99 Keating SM, Jacobs ES, Norris PJ. Soluble mediators of inflammation in HIV and their implications for therapeutics and vaccine development. Cytokine Growth Factor Rev 2012; 23: 193–206. 100 Nowak P, Barqasho B, Sönnerborg A. Elevated plasma levels of high mobility group box protein 1 in patients with HIV-1 infection. AIDS 2007; 21: 869–71. 101 Bjerk SM, Baker JV, Emery S, et al, and the INSIGHT SMART Study Group. Biomarkers and bacterial pneumonia risk in patients with treated HIV infection: a case-control study. PLoS one 2013; 8: e56249. 102 Vanham G, Penne L, Devalck J, et al. Decreased CD40 ligand induction in CD4 T cells and dysregulated IL-12 production during HIV infection. Clin Exp Immunol 1999; 117: 335–42. 103 Kelly-Scumpia KM, Scumpia PO, Weinstein JS, et al. B cells enhance early innate immune responses during bacterial sepsis. J Exp Med 2011; 208: 1673–82. 104 Kasten KR, Tschöp J, Adediran SG, Hildeman DA, Caldwell CC. T cells are potent early mediators of the host response to sepsis. Shock 2010; 34: 327–36. 105 Boomer JS, To K, Chang KC, et al. Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA 2011; 306: 2594–605. 106 Gougeon ML. To kill or be killed: how HIV exhausts the immune system. Cell Death Differ 2005; 12 (suppl 1): 845–54. 107 Piguet V, Schwartz O, Le Gall S, Trono D. The downregulation of CD4 and MHC-I by primate lentiviruses: a paradigm for the modulation of cell surface receptors. Immunol Rev 1999; 168: 51–63. 108 Daigneault M, De Silva TI, Bewley MA, et al. Monocytes regulate the mechanism of T-cell death by inducing Fas-mediated apoptosis during bacterial infection. PLoS Pathog 2012; 8: e1002814. 109 Klatt NR, Brenchley JM. Th17 cell dynamics in HIV infection. Curr Opin HIV AIDS 2010; 5: 135–40. 110 Leeansyah E, Ganesh A, Quigley MF, et al. Activation, exhaustion, and persistent decline of the antimicrobial MR1-restricted MAIT- cell population in chronic HIV-1 infection. Blood 2013; 121: 1124–35.
  • 13. www.thelancet.com/infection Vol 15 January 2015 107 Review 111 Cosgrove C, Ussher JE, Rauch A, et al. Early and nonreversible decrease of CD161++ /MAIT cells in HIV infection. Blood 2013; 121: 951–61. 112 Greathead L, Metcalf R, Gazzard B, Gotch F, Steel A, Kelleher P. CD8+/CD161++ mucosal-associated invariant T-cell levels in the colon are restored on long-term antiretroviral therapy and correlate with CD8+ T-cell immune activation. AIDS 2014; 28: 1690–92. 113 Gold MC, Lewinsohn DM. Co-dependents: MR1-restricted MAIT cells and their antimicrobial function. Nat Rev Microbiol 2013; 11: 14–19. 114 Rubino SJ, Geddes K, Girardin SE. Innate IL-17 and IL-22 responses to enteric bacterial pathogens. Trends Immunol 2012; 33: 112–18. 115 Zhang Z, Clarke TB, Weiser JN. Cellular effectors mediating Th17-dependent clearance of pneumococcal colonization in mice. J Clin Invest 2009; 119: 1899–909. 116 Catalfamo M, Wilhelm C, Tcheung L, et al. CD4 and CD8 T cell immune activation during chronic HIV infection: roles of homeostasis, HIV, type I IFN, and IL-7. J Immunol 2011; 186: 2106–16. 117 Sachdeva M, Fischl MA, Pahwa R, Sachdeva N, Pahwa S. Immune exhaustion occurs concomitantly with immune activation and decrease in regulatory T cells in viremic chronically HIV-1-infected patients. J Acquir Immune Defic Syndr 2010; 54: 447–54. 118 Larsson M, Shankar EM, Che KF, et al. Molecular signatures of T-cell inhibition in HIV-1 infection. Retrovirology 2013; 10: 31. 119 Champagne P, Ogg GS, King AS, et al. Skewed maturation of memory HIV-specific CD8 T lymphocytes. Nature 2001; 410: 106–11. 120 Appay V, Nixon DF, Donahoe SM, et al. HIV-specific CD8(+) T cells produce antiviral cytokines but are impaired in cytolytic function. J Exp Med 2000; 192: 63–75. 121 Andersson J, Behbahani H, Lieberman J, et al. Perforin is not co-expressed with granzyme A within cytotoxic granules in CD8 T lymphocytes present in lymphoid tissue during chronic HIV infection. AIDS 1999; 13: 1295–303. 122 Lange CG, Valdez H, Medvik K, Asaad R, Lederman MM. CD4+ T-lymphocyte nadir and the effect of highly active antiretroviral therapy on phenotypic and functional immune restoration in HIV-1 infection. Clin Immunol 2002; 102: 154–61. 123 Moir S, Fauci AS. Insights into B cells and HIV-specific B-cell responses in HIV-infected individuals. Immunol Rev 2013; 254: 207–24. 124 Hart M, Steel A, Clark SA, et al. Loss of discrete memory B cell subsets is associated with impaired immunization responses in HIV-1 infection and may be a risk factor for invasive pneumococcal disease. J Immunol 2007; 178: 8212–20. 125 Qiao X, He B, Chiu A, Knowles DM, Chadburn A, Cerutti A. Human immunodeficiency virus 1 Nef suppresses CD40- dependent immunoglobulin class switching in bystander B cells. Nat Immunol 2006; 7: 302–10. 126 Nielsen H, Kvinesdal B, Benfield TL, Lundgren JD, Konradsen HB. Rapid loss of specific antibodies after pneumococcal vaccination in patients with human immunodeficiency virus-1 infection. Scand J Infect Dis 1998; 30: 597–601. 127 Malaspina A, Moir S, Kottilil S, et al. Deleterious effect of HIV-1 plasma viremia on B cell costimulatory function. J Immunol 2003; 170: 5965–72. 128 Pensieroso S, Galli L, Nozza S, et al. B-cell subset alterations and correlated factors in HIV-1 infection. AIDS 2013; 27: 1209–17. 129 Moir S, Ho J, Malaspina A, et al. Evidence for HIV-associated B cell exhaustion in a dysfunctional memory B cell compartment in HIV-infected viremic individuals. J Exp Med 2008; 205: 1797–805. 130 Moir S, Buckner CM, Ho J, et al. B cells in early and chronic HIV infection: evidence for preservation of immune function associated with early initiation of antiretroviral therapy. Blood 2010; 116: 5571–79. 131 Jacobson MA, Khayam-Bashi H, Martin JN, Black D, Ng V. Effect of long-term highly active antiretroviral therapy in restoring HIV- induced abnormal B-lymphocyte function. J Acquir Immune Defic Syndr 2002; 31: 472–77. 132 Iwajomo OH, Finn A, Moons P, et al. Deteriorating pneumococcal-specific B-cell memory in minimally symptomatic African children with HIV infection. J Infect Dis 2011; 204: 534–43. 133 Moja P, Jalil A, Quesnel A, et al. Humoral immune response within the lung in HIV-1 infection. Clin Exp Immunol 1997; 110: 341–48. 134 Gordon SB, Miller DE, Day RB, et al. Pulmonary immunoglobulin responses to StreptococcuS pneumoniae are altered but not reduced in human immunodeficiency virus-infected Malawian adults. J Infect Dis 2003; 188: 666–70. 135 Takahashi H, Oishi K, Yoshimine H, et al. Decreased serum opsonic activity against StreptococcuS pneumoniae in human immunodeficiency virus-infected Ugandan adults. Clin Infect Dis 2003; 37: 1534–40. 136 MacLennan CA, Gilchrist JJ, Gordon MA, et al. Dysregulated humoral immunity to nontyphoidal Salmonella in HIV-infected African adults. Science 2010; 328: 508–12. 137 Matta F, Yaekoub AY, Stein PD. Human immunodeficiency virus infection and risk of venous thromboembolism. Am J Med Sci 2008; 336: 402–06. 138 Jong E, Louw S, van Gorp EC, Meijers JC, ten Cate H, Jacobson BF. The effect of initiating combined antiretroviral therapy on endothelial cell activation and coagulation markers in South African HIV-infected individuals. Thromb Haemost 2010; 104: 1228–34. 139 Koppel K, Bratt G, Schulman S, Bylund H, Sandström E. Hypofibrinolytic state in HIV-1-infected patients treated with protease inhibitor-containing highly active antiretroviral therapy. J Acquir Immune Defic Syndr 2002; 29: 441–49. 140 Holme PA, Müller F, Solum NO, Brosstad F, Frøland SS, Aukrust P. Enhanced activation of platelets with abnormal release of RANTES in human immunodeficiency virus type 1 infection. FASEB J 1998; 12: 79–89. 141 Mayne E, Funderburg NT, Sieg SF, et al. Increased platelet and microparticle activation in HIV infection: upregulation of P-selectin and tissue factor expression. J Acquir Immune Defic Syndr 2012; 59: 340–46. 142 Funderburg NT, Mayne E, Sieg SF, et al. Increased tissue factor expression on circulating monocytes in chronic HIV infection: relationship to in vivo coagulation and immune activation. Blood 2010; 115: 161–67. 143 Lafeuillade A, Alessi MC, Poizot-Martin I, et al. Endothelial cell dysfunction in HIV infection. J Acquir Immune Defic Syndr 1992; 5: 127–31. 144 Ross AC, Armentrout R, O’Riordan MA, et al. Endothelial activation markers are linked to HIV status and are independent of antiretroviral therapy and lipoatrophy. J Acquir Immune Defic Syndr 2008; 49: 499–506. 145 Solages A, Vita JA, Thornton DJ, et al. Endothelial function in HIV-infected persons. Clin Infect Dis 2006; 42: 1325–32. 146 Zietz C, Hotz B, Stürzl M, Rauch E, Penning R, Löhrs U. Aortic endothelium in HIV-1 infection: chronic injury, activation, and increased leukocyte adherence. Am J Pathol 1996; 149: 1887–98. 147 Joshi VV, Pawel B, Connor E, et al. Arteriopathy in children with acquired immune deficiency syndrome. Pediatr Pathol 1987; 7: 261–75. 148 Mankhambo LA, Banda DL, Jeffers G, et al, and the IPD Study Group. The role of angiogenic factors in predicting clinical outcome in severe bacterial infection in Malawian children. Crit Care Med 2010; 14: R91. 149 de Larrañaga GF, Bocassi AR, Puga LM, Alonso BS, Benetucci JA. Endothelial markers and HIV infection in the era of highly active antiretroviral treatment. Thromb Res 2003; 110: 93–98. 150 de Gaetano Donati K, Rabagliati R, Tumbarello M, et al. Increased soluble markers of endothelial dysfunction in HIV-positive patients under highly active antiretroviral therapy. AIDS 2003; 17: 765–68. 151 Rittirsch D, Redl H, Huber-Lang M. Role of complement in multiorgan failure. Clin Dev Immunol 2012; 2012: 962927. 152 Huber G, Bánki Z, Lengauer S, Stoiber H. Emerging role for complement in HIV infection. Curr Opin HIV AIDS 2011; 6: 419–26. 153 Yefenof E, Asjö B, Klein E. Alternative complement pathway activation by HIV infected cells: C3 fixation does not lead to complement lysis but enhances NK sensitivity. Int Immunol 1991; 3: 395–401.
  • 14. 108 www.thelancet.com/infection Vol 15 January 2015 Review 154 Dellinger RP, Levy MM, Rhodes A, et al, and the Surviving Sepsis Campaign Guidelines Committee including the Pediatric Subgroup. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med 2013; 41: 580–637. 155 Hutchins NA, Unsinger J, Hotchkiss RS, Ayala A. The new normal: immunomodulatory agents against sepsis immune suppression. Trends Mol Med 2014; 20: 224–33. 156 Fagard C, Le Braz M, Günthard H, et al, and the Swiss HIV Cohort Study. A controlled trial of granulocyte macrophage-colony stimulating factor during interruption of HAART. AIDS 2003; 17: 1487–92. 157 Porichis F, Hart MG, Zupkosky J, et al. Differential impact of PD- 1 and/or interleukin-10 blockade on HIV-1-specific CD4 T cell and antigen-presenting cell functions. J Virol 2014; 88: 2508–18. 158 Vanham G, Van Gulck E. Can immunotherapy be useful as a “functional cure” for infection with Human Immunodeficiency Virus-1? Retrovirology 2012; 9: 72. 159 Zhou H, Ji X, Wu Y, et al. A dual-role of Gu-4 in suppressing HMGB1 secretion and blocking HMGB1 pro-inflammatory activity during inflammation. PLoS one 2014; 9: e89634. 160 Vincent JL, Ramesh MK, Ernest D, et al. A randomized, double- blind, placebo-controlled, Phase 2b study to evaluate the safety and efficacy of recombinant human soluble thrombomodulin, ART-123, in patients with sepsis and suspected disseminated intravascular coagulation. Crit Care Med 2013; 41: 2069–79.