SlideShare a Scribd company logo
1 of 38
Download to read offline
A
B	
  
30μm	
  
30μm	
  
Figure	
  1	
  Representa5ve	
  
epifluorescence	
  graphs	
  of	
  cultured	
  rat	
  
DRG	
  neurons	
  before	
  (A)	
  and	
  aCer	
  (B)	
  
applica5on	
  of	
  100nM	
  capsaicin.	
  
Arrows,	
  Fura-­‐2-­‐loaded	
  cells	
  exhibi5ng	
  
Ca2+	
  	
  mobiliza5on	
  in	
  response	
  to	
  
capsaicin.	
  (Scale	
  bars,	
  30μm.)	
  
0	
  
0.5	
  
1	
  
1.5	
  
2	
  
2.5	
  
3	
  
3.5	
  
4	
  
0	
   150	
   300	
   450	
   600	
   750	
   900	
   1050	
   1200	
  
(Sec)	
  
0	
  
0.5	
  
1	
  
1.5	
  
2	
  
2.5	
  
0	
   150	
   300	
   450	
   600	
  
(Sec)	
  
∆Ra5o	
  340/380	
  ∆Ra5o	
  340/380	
  
Figure	
  2	
  Representa5ve	
  traces	
  of	
  
capsaicin	
  evoked	
  calcium	
  
responses	
  in	
  rat	
  DRG	
  neurons	
  
(Blue),	
  enhanced	
  by	
  laminectomy	
  
(Red).	
  	
  Intrathecal	
  pretreatment	
  
with	
  the	
  TRPV1	
  agonist	
  RTX	
  
a^enuated	
  laminectomy	
  induced	
  
enhancement	
  (Green).	
  	
  
Figure	
  3	
  	
  On	
  POD1,	
  potassium	
  chloride	
  evoked	
  calcium	
  transients	
  in	
  rat	
  DRG	
  
neurons	
  as	
  measured	
  by	
  the	
  ra5o	
  difference	
  between	
  the	
  peak	
  ra5o	
  and	
  
baseline	
  ra5o	
  (∆Ra5o	
  340/380).	
  Data	
  are	
  presented	
  as	
  mean±SEM	
  of	
  	
  ∆Ra5o	
  
340/380.	
  ***p<0.001,	
  **p<0.01,	
  n=50-­‐80	
  cells	
  from	
  4-­‐6	
  rats	
  each	
  group.	
  
Figure	
  4	
  On	
  POD1,	
  dura5on	
  (seconds)	
  of	
  recovery	
  to	
  80%	
  of	
  the	
  baseline	
  of	
  K+	
  evoked	
  Ca2+	
  
transients	
  in	
  rats	
  DRG	
  neurons.	
  Data	
  are	
  presented	
  as	
  mean±SEM	
  of	
  the	
  5me	
  dura5on	
  to	
  80%	
  
recovery	
  of	
  baseline	
  of	
  ∆Ra5o	
  340/380.	
  ***p<0.001,	
  **p<0.01,	
  n=	
  50-­‐80	
  cells	
  from	
  4-­‐6	
  rats	
  each	
  
group.	
  
Figure	
  5	
  On	
  POD1,	
  capsaicin	
  evoked	
  calcium	
  transients	
  in	
  rat	
  DRG	
  neurons	
  as	
  
measured	
  by	
  the	
  ∆Ra5o	
  340/380.	
  Data	
  are	
  presented	
  as	
  mean±SEM	
  of	
  	
  ∆Ra5o	
  
340/380.	
  ***p<0.001,	
  *p<0.05,	
  n=45-­‐70	
  cells	
  from	
  4-­‐6	
  rats	
  each	
  group.	
  
Figure	
  6	
  On	
  POD1,	
  dura5on	
  (seconds)	
  of	
  recovery	
  to	
  80%	
  of	
  the	
  baseline	
  of	
  
capsaicin	
  evoked	
  Ca2+	
  transients	
  in	
  rats	
  DRG	
  neurons.	
  Data	
  are	
  presented	
  as	
  
mean±SEM	
  of	
  the	
  5me	
  dura5on	
  to	
  80%	
  recovery	
  of	
  baseline	
  of	
  ∆Ra5o	
  340/380.	
  
***p<0.001,	
  *p<0.05,	
  n=	
  45-­‐70	
  cells	
  from	
  4-­‐6	
  rats	
  each	
  group.	
  
Figure	
  7	
  On	
  POD1,	
  percentage	
  of	
  capsaicin	
  responsive	
  DRG	
  neurons.	
  Data	
  are	
  
presented	
  as	
  mean±SEM	
  of	
  the	
  percentage	
  of	
  neurons	
  responsive	
  to	
  capsaicin	
  in	
  
the	
  whole	
  DRG.	
  ***p<0.001,	
  *p<0.05,	
  n=	
  45-­‐70	
  cells	
  from	
  4-­‐6	
  rats	
  each	
  group.	
  
Figure	
  8	
  On	
  POD7,	
  potassium	
  chloride	
  evoked	
  calcium	
  transients	
  in	
  rat	
  DRG	
  
neurons	
  as	
  measured	
  by	
  the	
  ra5o	
  difference	
  between	
  the	
  peak	
  ra5o	
  and	
  baseline	
  
ra5o	
  (∆Ra5o	
  340/380).	
  Data	
  are	
  presented	
  as	
  mean±SEM	
  of	
  	
  ∆Ra5o	
  340/380.	
  
**p<0.01,	
  n=45-­‐70	
  cells	
  from	
  4-­‐6	
  rats	
  each	
  group.	
  
	
  Figure	
  9	
  On	
  POD7,	
  dura5on	
  (seconds)	
  of	
  recovery	
  to	
  80%	
  of	
  the	
  baseline	
  of	
  K+	
  
evoked	
  Ca2+	
  transients	
  in	
  rats	
  DRG	
  neurons.	
  Data	
  are	
  presented	
  as	
  mean±SEM	
  of	
  
the	
  5me	
  dura5on	
  to	
  80%	
  recovery	
  of	
  baseline	
  of	
  ∆Ra5o	
  340/380.	
  **p<0.01,	
  n=	
  
45-­‐70	
  cells	
  from	
  4-­‐6	
  rats	
  each	
  group.	
  
	
  
la
m
in
e
c
to
m
y
s
h
a
m
s
u
rg
e
ry
0 .0
0 .5
1 .0
1 .5
DRatio340/380
la
m
in
e
c
to
m
y
s
h
a
m
s
u
rg
e
ry
0
5 0
1 0 0
1 5 0
t80%(seconds)
Figure	
  10	
  On	
  POD7,	
  capsaicin	
  evoked	
  calcium	
  transients	
  in	
  rat	
  DRG	
  neurons	
  as	
  
measured	
  by	
  the	
  ra5o	
  difference	
  between	
  the	
  peak	
  ra5o	
  and	
  baseline	
  ra5o	
  
(∆Ra5o	
  340/380).	
  Data	
  are	
  presented	
  as	
  mean±SEM	
  of	
  	
  ∆Ra5o	
  340/380.	
  
***p<0.001,	
  n=40-­‐60	
  cells	
  from	
  4-­‐6	
  rats	
  each	
  group.	
  
	
  Figure	
  11	
  On	
  POD7,	
  dura5on	
  (seconds)	
  of	
  recovery	
  to	
  80%	
  of	
  the	
  baseline	
  of	
  K+	
  evoked	
  
Ca2+	
  transients	
  in	
  rats	
  DRG	
  neurons.	
  Data	
  are	
  presented	
  as	
  mean±SEM	
  of	
  the	
  5me	
  
dura5on	
  to	
  80%	
  recovery	
  of	
  baseline	
  of	
  ∆Ra5o	
  340/380.	
  ***P<0.001,	
  **p<0.01,	
  n=	
  40-­‐60	
  
cells	
  from	
  4-­‐6	
  rats	
  each	
  group.	
  
	
  
Figure	
  12	
  On	
  POD7,	
  percentage	
  of	
  capsaicin	
  responsive	
  DRG	
  neurons.	
  Data	
  are	
  
presented	
  as	
  mean±SEM	
  of	
  the	
  percentage	
  of	
  neurons	
  responsive	
  to	
  capsaicin	
  in	
  the	
  
whole	
  DRG.	
  ***p<0.001,	
  n=	
  40-­‐60	
  cells	
  from	
  4-­‐6	
  rats	
  each	
  group.	
  
	
  
On	
  pod6	
  7	
  there	
  is	
  a	
  sig	
  diff	
  between	
  RTX	
  and	
  no	
  drug	
  laminectomy	
  in	
  running	
  
distance	
  (two	
  way	
  anova,	
  for	
  mul5ple	
  t-­‐	
  test,	
  day	
  3,4,5,6,7	
  is	
  different.	
  P=0.016,	
  
0.0013,0.0013,0.0002,0.0004).	
  On	
  pod0,	
  there	
  is	
  a	
  diff	
  between	
  RTX	
  and	
  no	
  drug	
  
in	
  maximum	
  speed(two	
  way	
  anova,mul5ple	
  t	
  test,	
  day	
  0,1,2,	
  
p=0.0003,0.01,0.02).	
  there	
  is	
  no	
  diff	
  between	
  the	
  2	
  group	
  in	
  accelera5on.	
  
there	
  is	
  a	
  sig	
  diff	
  between	
  RTX	
  and	
  no	
  drug	
  laminectomy	
  in	
  fine	
  motor	
  
movement	
  (two	
  way	
  anova,	
  for	
  mul5ple	
  t-­‐	
  test,	
  day	
  1,2	
  is	
  different.	
  P=0.014,	
  
0.026).	
  there	
  is	
  a	
  diff	
  between	
  RTX	
  and	
  no	
  drug	
  in	
  rearing(two	
  way	
  
anova,mul5ple	
  t	
  test,	
  day	
  1,	
  p=0.008).	
  there	
  is	
  no	
  diff	
  between	
  the	
  2	
  group	
  in	
  
accelera5on.	
  
Figure	
  13	
  On	
  baseline,	
  post	
  RTX	
  injec5on	
  day	
  7,	
  post	
  RTX	
  
injec5on	
  day	
  14,	
  thermal	
  pain	
  withdrawal	
  threshold	
  with	
  a	
  
cutoff	
  of	
  25	
  seconds.	
  *p<0.05,	
  n=4	
  rats	
  each	
  group.	
  
Prolonged and enhanced rodent intracellular Ca2+ signal in
dorsal root ganglion after laminectomy was shortened and down
regulated by intrathecal resiniferatoxin
Abstract
Background: postoperative pain is a challenging problem in clinic, compromising
quality of life and leading to extended hospital stay and economic burden and could
develop into chronic pain. Patients report significant pain and hypersensitivity
postoperatively. This study examined the capsaicin and high potassium induced
intracellular calcium response in cultured adult rat DRG neurons on postoperative day
1 and postoperative day 7, and examined the effects of resiniferatoxin treatment on
neuronal intracellular calcium responses and rats behavior including wheel running and
spontaneous home cage activities.
Results: Calcium imaging showed that laminectomy resulted in prolonged duration and
elevated amplitude of high potassium and capsaicin induced DRG neuron intracellular
calcium responses. Also the surgery led to higher responsive ratio in DRG populations,
compared to controls; this was reduced by intrathecal administration of 200ng
resiniferatoxin. Laminectomy resulted in decreased wheel running distance, maximum
speed , home cage rearing and fine movements. Resiniferatoxin partially reversed the
effects of laminectomy on wheel running and home cage spontaneous activities. It also
extended the heat response in hind paw after intrathecal injection.
Conclusions: Laminectomu induces TRP sensitization and higher response to high
potassium in DRG neurons and leads to reduced wheel running and spontaneous
activities in home cage. These effects may be mitigated by preoperative treatment with
TRPV1 agonist resiniferatoxin which could lead to desensitization of neurons, increase
wheel running and alleviate spontaneous pain.
Introduction
A large body of evidence point to the importtant role of TrpV1, the capsaicin receptor, in
sensation of noxious heat (Caterina et al., 1997). TrpV1is also believed to play a role in
increased
pain sensitivity following inflammation at least in part because inflammation induced
phosphorylation of TrpV1 results in increased sensitivity of this ion channel to
temperature (Bhave et al., 2002; Cesare et al., 1999; Nilius et al., 2005; Numazaki et
al., 2002). Attention has focused on TrpV1 and TrpV1 cells for their role in pain
sensation and as targets for pain therapies. Notably, TrpV1 antagonists have been
shown to be efficacious in animal models of chronic pain(Szallasi et al.,2007;Wong
andGavva,2009) but, as yet, have not been exploited in human pain management.
An alternate strategy for pain management involves ablation of TrpV1-expressing cells
or TrpV1 fibers. This approach has a long history: more than 25 years ago, capsaicin
injection was reported to dramatically decrease the level of inflammatory mediators
(e.g., substance P) and pain sensation for the life of animals (Jancso et al., 1977; Nagy
et al., 1981; Yaksh et al., 1979). More recently, the potent TrpV1 agonist, resiniferotoxin
(RTX) has been demonstrated to kill TrpV1-expressing neurons by affecting long-term
Ca2+ entry (Olah eal., 2001). RTX has also been used therapeutically in animals and
has been reported to alleviate chronic pain with no effects on discriminative touch or
proprioception (Brown et al., 2005; Karai et al., 2004; Neubert et al., 2003). In a rat
model, RTX was shown to eliminate many TrpV1 neurons, dramatically decrease
responses to capsaicin and thermal pain (Karai et al., 2004), and affect
mechanosensation (Tender et al., 2008).
Mishra et al 1
carried out a detailed investigation into the efficacy of RTX in ablating
sensory neurons and demonstrate that although a large subset of TrpV1neurons are
killed by a single injection, other TrpV1 cells are not affected even by repeated RTX
treatments. RTX reduces but does not eliminate capsaicin responses and thermal
sensation of normal mice.
We hypothesized that intrathecal application of RTX may block the DRB neurons
temporarily and reduce the pain behavior. RTX is supposed to work on trpv1 positive
small diameter neurons and leave intact the large diameter axons responsible for
proprioceptive and somatic sensations.The intrathecal approach is proposed to broadly
affect the DRGs surrounding the L4, L5 and their corresponding dermatome including
the laminectomy site to mediate a selective antinociceptive effects. By modifying the
trpv1 positive DRG neurons, this treatment can help relieve different clinical pain
including the postoperative pain.
Here, we set out to determine the function of TrpV1 cells and explored the use of RTX
as a means to affect the intracellular calcium response to capsaicin and hight potassium
stimulations. And to see if it can selectively eliminate trpv1 positive neurons in rats. The
primary aim of this study was to determine whether incisional pain produced a higher
calcium response in DRG neurons and how long it will last. The other aim of the study
was to determine if intrathecal RTX will promote the reduction of calcium response in
these DRG neurons after laminectomy and to check the effect of RTX on the hind paw
thermal response and the effect of RTX on wheel running and rodent spontaneous pain
behavior after intrathecal application. Low doses of RTX (100-250ng) has been proved
to be effective when given perineurally2
, in single ganglionic injection or intrathecally3
.
The minimus effective doses should be determined to avoid the po tential side effects
considering the clinical settings. So this study used the 200ng RTX to block multiple
ganglia by applying intrathecally.
Materials and Methods
Animals
Experiments were conducted in adult male Sprague-Dawley rats (Harlan,Indianapolis,
IN, USA) weighing 225-250 g. Animals were housed in groups of 2, maintained in a 12-
hour light-dark cycle with a temperature-controlled environment and given food and
water ad libitum. All procedures used in these experiments were reviewed and
approved by the Institutional Animal Care and Use Committee at the University of Iowa.
Animals were cared for and used in accordance with guidelines of the Committee for
Research and Ethical Issues of the IASP4
.
Chemicals and reagents
1,1'-Dioleyl-3,3,3',3'-Tetramethylindocarbocyanine methanesulfonate (DiI), Fura-2
acetoxymethyl (AM) ester and Pluronic F-127 (stock concentration 20%) were
purchased from Invitrogen (Carlsbad, CA, USA). Concentrations were based on results
from previous studies5
. DiI was dissolved in DMSO (25 mg/ml). Fura-2 AM (2.5µm) was
dissolved in 1mM dimethyl sulfoxide and stored at -20°C. When ready to use, the
Fura-2AM was diluted in bath solution (in mM: 130 NaCl, 5 KCl, 2 CaCl2, 1 MgCl2, 10
Hepes, 10 glucose, and pH adjusted to 7.4, and osmolality adjusted with sucrose to
310-325 mOsm), together with Pluronic F-127 (0.025%) immediately before use.
Resiniferatoxin (RTX), (Sigma Aldrich, St Louis, MO, USA) was dissolved in 10/10/80%
mix of ethanol, Tween-80, and saline. Capsaicin (Sigma, St. Louis, MO, USA) was
diluted to a final concentration of 1mM in 1.5% Tween-80, 1.5% alcohol and 97% saline.
Capsaicin (100nM) was diluted in normal bath solution immediately before use. All other
compounds were dissolved in normal bath solution directly. For obtaining high-
potassium extracellular solution (K+
, 30mM), the K+
in normal bath solution was adjusted
to 30 mM by iso-osmotic replacement of Na+
.
Labeling DRG neurons
Prior to tissue harvest (>5 days), sensory neurons innervating the lower back region
were labelled with the retrograde tracer DiI. General anesthesia was induced with
isoflurane (Abbott Laboratories, North Chicago, IL, USA) in room air and maintained at
1.5%, DiI solution (8 µl) was injected onto the right L4 and L5 laminae and surrounding
muscles (4 µl ×2) by positioning the needle 2mm away from the midline (the L4 and L5
spinous processes) with a 30 g injection needle.	
  	
  
Intrathecal injection of RTX or Saline
Under 1.5% isoflurane anesthesia, each rat in the RTX group received intrathecal
(between the L4 and L5 vertebrae) injection of 20 µl RTX (200ng in 20 µl saline) using a
Hamilton syringe and a 25-gauge needle. Each rat in saline group received injection of
20 µl saline.
Behavioral pain tests
Rats in the RTX group were tested on baseline, post injection day 7 and post injection
day 14. For withdrawal latency to radiant heat stimuli, unrestrained rats were individually
placed onto a 3-mm thick glass surface (40 × 40 cm) under a plastic chamber (21 ×
27×15 cm) for approximately 15 minutes of acclimation. A focused radiant heat source
was applied from underneath the glass floor onto the center of hind paw. The latency to
evoke withdrawal was determined with a cut-off value of 30 seconds to prevent radiant
heat evoked injury to the hindpaw. Each rat was tested 3 times, with a 5-minute interval
between tests. The mean paw withdrawal latency was determined from 3 tests.
Guarding pain was performed before withdrawal tests to avoid the potential influence of
withdrawal responses on guarding pain.
Wheel running and home cage locomotion tests
Rats were placed individually in cages fitted with exercise wheels (Intellibio Activity
Wheel, Seichamps, France). The rats were housed individually and allow to run on the
exercise wheels with a computer that recorded the distance, speed, acceleration,
revolutions and time (in one hour bins). A stable baseline was achieved after 14 days of
running wheel exposure. The last 3 days were averaged to obtain the baseline. Data
was recorded daily for 14 days. The distance traveled by the rat, acceleration, maximum
speed, total motor movement, and rearing activity of rats became stable after 2 weeks
habituation with this experimental protocol. The effects of a treatment on each rat were
analyzed as percentage of the distance traveled during the baseline.
Home Cage Activity Activity during the dark cycle (period when rats are active) after
surgery was monitored. After acclimatization to individual home cages (17 x 5 x 18 inch)
for 3 days, the cages were inserted into a SmartFrame Cage Rack Motor Monitor
System (Kinder scientific, Poway, CA). Two grids of 7x15 infrared beams surrounded
the cage; one was placed 6 cm above the floor to record total locomotor activity as total
beam interruptions. The second series of beams was placed 18 cm above the cage
floor and count rearing, when the rat stood on its hindpaws. Beam breaks were counted
to record total activity and rearing. The data was then transmitted to a host computer
and collected in 1-hour bins. The last 3 days were averaged as the baseline, and then
rats would undergo sham surgery or full laminectomy. We used these monitors to
examine hypermobility by acute pain stimuli.
Laminectomy Procedure
After the induction of general anesthesia with isoflurane in room air and maintained with
1.5% isoflurane. The surgery was performed with spontaneous ventilation through a
nose cone. Each animal was shaved and sterilized with povidone iodine at the incision
site. A 4cm midline longitudinal incision is made with a # 11 blade through the skin
down to the fascia between L3-L6. Blunt dissection was continued through the incision
into the muscle (removing muscle away from bone) to further divide and retracts the
muscle. The posterior bony spine was exposed and L4-L6 spinous processes removed.
The bone between L4-L5 was removed using a battery-powered drill. The dura was
exposed without breaching it. After surgery, the wound was closed in layers; the muscle
was closed with 4 sutures, followed by fascia using absorbable 3-0 vicryl suture
(Ethicon, San Angelo, TX, USA). The skin was closed with 4 sutures of 3-0 nylon
(Ethicon, San Angelo, TX, USA).
Cell Culture
Rats were euthanized with 100% carbon dioxide and the right-sided L1-L3 DRG were
surgically removed and desheathed in ice-cold Hanks balanced salt solution (HBSS)
(Gibco, Gaithersburg, MD, USA). DRGs were incubated 20 min at 37°C in 3 ml HBSS
containing 0.125% papain (Worthington, Lakewood, NJ, USA) and then centrifuged at
800 g for 1 min. The papain solution was removed and replaced and incubated for 20
min with 3ml HBSS containing 10 mg collagenase IV (Worthington, Lakewood, NJ,
USA) and 14mg dispase (Invitrogen, Carlsbad, CA, USA). DRGs were then
centrifuged at ∼800 g for 1 min, resuspended with culture media F12 (Gibco,
Gaithersburg, MD, USA). DRGs were mechanically dissociated in this solution with fire-
polished Pasteur pipettes and then plated onto glass coverslips, previously coated by a
solution of 0.1 mg/ml poly-D-lysine (Sigma Aldrich, St Louis, MO,USA) and 5 µg/ml
mouse laminin (BD Bioscience, Bedford, CA, USA). A 0.5-mm-thick silicone ring with a
4-mm-diameter opening sealed to the surface of each well reduced the plating area in
each well. The cells were incubated at 37°C, in 5% CO2 for 2 h. After 2 h, the wells were
filled with 2ml F12 media (Gibco, Gaithersburg, MD, USA) containing 10%(v/v) Fetal
Bovine Serum (FBS), 50 U/ml penicillin , 50 µg/ml streptomycin, and 50ng/ml nerve
growth factor (NGF) (Sigma Aldrich, St Louis, MO,USA), and incubated overnight.
Subclassification of DRG neurons
Neurons were divided into small (<30 µm), and large (≥30 µm) based on cell body
diameter as measure with a calibrated eyepiece reticule. Only neurons with visible
nucleus are used. Neurons with a small cell body diameter are likely to be nociceptive
afferents, while those with a large cell body diameter are likely to be non-nociceptive
afferents6
.
Intracellular Ca2+
measurements
All experiments were performed within 24h of tissue harvest. DRG neurons were loaded
with the calcium indicator dye, Fura-2 AM ester (2.5µM) along with 0.025% Pluronic for
20 min at room temperature as previously described 5
. Following fura-2 loading, DRG
neurons were placed in a recording chamber mounted on the stage of an inverted IX-71
microscope (Olympus IX71, Japan) and were continuously perfused for 5 minutes with
normal bath solution before the experiment began. Fluorescence data were acquired on
computer running Slidebook 5.0 software (Molecular Devices, Sunnyvale, CA, USA) at
20°C. Only cells with DiI label were used for calcium imaging. Fluorescence was
alternately excited at 340nm and 380nm using the Lambda DG-4 filter changer (Sutter
Instruments, Novato, CA, USA) via a 10x (Numerical Aperture NA=0.30) or 20x
(NA=0.70) objectives. Emitted fluorescence was collected at 510nm using an ORCA-
03G cooled digital CCD camera (Hamamatsu Photonics, Japan). Pairs of images were
sampled at 1-3 Hz. For each field, an average of 20-40 cells for the 20x objective were
tracked in real time. The ratio of fluorescence emission (510 nm) in response to 340/380
nm excitation was acquired at 1-3 Hz during drug applications. All drugs were applied
via a gravity-fed chamber perfusion system. Cells were depolarized by capsaicin (100
nM, 30 seconds) or K+
(30 mM, 30 seconds). Cells not responsive to K+
were excluded.
Two properties of calcium influx were measured: peak ratio (340/380 nm) of the Fura-2
signal, and duration of the Fura-2 signal (calculated as time to 80% of baseline value).
In case of capsaicin depolarization, the percentages of cells responsive to capsaicin
were also compared between groups.
Statistical analysis
Data are expressed as mean± standard error of the mean (SEM) unless otherwise
stated. The “n” for each group refers to the number of neurons. Statistical significance
was determined using Student’s t-test, or ANOVA when appropriate followed by Tukey’s
post-hoc comparisons (GraphPad Prism software, San Diego, CA, USA). Differences at
the P<0.05 level were considered statistically significant.
Results
To assess the effect of laminectomy on the DRG neurons’ intracellular calcium
transients’ responses to K+
or capsaicin induced depolarization, Fura-2 Ca2+
imaging
experiments were performed on DRG neurons that were transiently depolarized with
30nM KCl or 100nM capsaicin. We only analyzed the L1-L3 DRG DiI+ neurons which
were supposed to represent the neurons innervating the laminectomy area at the lower
back region 7
. And only the calcium transients of the potassium responsive neurons
were analyzed.
On POD1, laminectomy potentiates K+ evoked Ca2+ transients in DRG neurons
and RTX attenuated the Ca2+ transients enhancement induced by laminectomy.
The magnitudes of K+ evoked Ca2+ transient were analyzed using the change of peak
ratio to baseline. And the decay was measured as time from onset to return to 20% of
peak (T80). Results of this analysis for DiI+ neurons revealed significant effects
associated with laminectomy (P<0.001). When using K+ to depolarize the DRG
neurons, both the magnitude and the decay of Ca2+ transients in DRG neurons were
enhanced by laminectomy. Laminectomy induced higher amplitude (laminectomy
1.70±0.07vs. sham 1.26±0.09, ***p<0.001, Fig.1) and longer decay (laminectomy
269.21±34.98 vs. sham 137.6±15.55, ***p<0.001, Fig.2) when compared with sham
group. Laminectomy significantly enhanced the Ca2+ transients evoked by 30nM KCl
depolarization and attenuated the recovery from depolarization evoked by 30nM KCl.To
determine whether RTX can have effect on DRG neurons’ K+ evoked Ca2+ transients,
we administered 200ng RTX intrathecally at L4 and L5 level 7 days before
laminectomy.Post-hoc (Tukey)analysis indicated that intrathecal 200ng RTX
pretreatment reduced the K+ evoked calcium flux in laminectomy group. (laminectomy
1.70±0.07, vs. RTX and laminectomy 1.14±0.07, *** p<0.001, n= 46 vs 31 cells, 6 rats,
Fig. 1), while the decay of the K+ evoked Ca2+ transient was shortened by RTX
pretreatment (laminectomy 269.2±34.98, vs. RTX and laminectomy 82.43±11.12, ***
p<0.001, n= 25 vs 21cells, 6 rats, ***P<0.001, Fig. 2). Thus, potassium-depolarization
mediated calcium mobilization in laminectomy group was drastically reduced in the
presence of RTX. On POD1, RTX also can reduce the amplitude of calcium flux in sham
rat DRG neurons. (sham 1.26±0.09, vs. RTX and sham 0.88±0.07, **p<0.01, n= 45 vs
44cells, 6 rats, Fig.1). and shortened the duration. (sham 137.7±15.55, vs. RTX and
sham 74.46±8.29, *p<0.05, n= 45,37 cells, 6 rats, Fig.2). Therefore, on POD1, in both
laminectomy rats and sham rats, RTX reduced the calcium response to KCl , either by
reducing the amplitude of calcium peak or by shortening the decay of the transient.
On POD1, capsaicin evoked Ca2+ transients in DRG neurons was also potentiated
by laminectomy, which was attenuated by intrathecal RTX.
Laminectomy induced higher amplitude when compared with sham group (laminectomy
2.60±0.14 vs. sham 2.03±0.14, *p<0.05, Fig. 3 Post hoc analysis (Tukey test)) and slow
the recovery of transients in small diameter neurons (laminectomy 986.10±116.10, vs.
sham 511.31±78.65, ***P<0.001, Fig. 4). And there is also a significant increase in the
percentage of capsaicin responsive neurons in the DRG populations induced by
laminectomy (laminectomy 75.20±3.79% vs. sham 63.67±1.93%, *P<0.05, Fig. 5).
As observed RTX effect on potassium induced calcium transients, there was also a
statistically significant effect of pre-surgery RTX in both the magnitude (laminectomy
2.60±0.14, vs. RTX and laminectomy 1.52±0.16, *** p<0.001, n= 40 vs 34 cells, 6 rats,
Fig. 3)and decay (laminectomy 986.10±116.10, vs. RTX and laminectomy
378.70±69.18, *** p<0.001, n= 32 vs 27 cells, 6 rats, Fig. 4) of the capsaicin evoked
Ca2+ transient in small DiI+ DRG neurons. In addition, RTX decreased the percentage
of capsaicin responsive DRG (laminectomy 75.20±3.79% vs. RTX 51.00±3.67%,
***p<0.001, Fig. 5). It also decreased the percentage of capsaicin responsive DRG in
sham rats (sham 63.67±1.93% vs. RTX and sham 33.10±1.78%, ****P<0.0001).
On POD1, in both laminectomy rats and sham rats, RTX reduced the calcium response
to capsaicin, either by completely eliminating the transient or by reducing the amplitude
of calcium peak or by shortening the decay of the transient.
On POD7, laminectomy did not produce significant changes in K+ evoked Ca2+
transients in DRG neurons either in amplitude (laminectomy 1.04±0.11vs. sham
1.18±0.08, p>0.05,n= 23 vs 40 cells, 4 rats Fig. 6) or decay(laminectomy 108.70±26.31
vs. sham 116.2±12.79, p>0.05, Fig. 7).
On POD7, laminectomy extended the decay of capsaicin evoked Ca2+ transients.
And RTX attenuated the laminectomy enhanced capsaicin response in DRG
neurons
On POD7, even the magnitude of capsaicin evoked Ca2+ transients did not differ
between laminectomy group and sham group(laminectomy 2.19±0.18 vs. sham
1.77±0.15,p>0.05, n=20 vs 38 cells, 4 rats each group Fig. 8) although RTX decrease
the amplitude both in laminectomy rats (laminectomy 2.19±0.18, vs. RTX and
laminectomy 1.21±0.12, n=20 vs 37 cells , 4 rats each group,*** p<0.001, Fig. 8) and
sham rats(sham 1.77±0.15% vs RTX and sham 0.85±0.09%, n=38 vs 30 cells, 4 rats
each group, ***P<0.001, Fig. 8).
However there was still increased decay of capsaicin evoked Ca2+ transients in
laminectomy group (laminectomy 1044±149 vs sham 414±44.76 seconds, n= 25vs 33
cells, 4 rats each group, ***P<0.001, Fig. 9), and RTX shortened the decay in both
laminectomy rat (laminectomy 1044.0±149.3 vs RTX and laminectomy 188.6±33.68
seconds, n=25 vs 26 cells, 4 rats each group, ****P<0.0001, Fig. 9) and sham
rats(sham 414±44.76% vs RTX and sham 161.2±11.89%, n=33 vs 35 cells, 4 rats each
group, *P<0.05, Fig. 9).
On POD7, there was no significant difference in terms of the percentage of capsaicin
responsive neurons between the sham group and laminectomy group (laminectomy
71.75±6.96% vs. sham 59.51±2.82%, 4 rats each group, p>0.05, Fig. 10), even when
RTX still lowered the percentage of capsaicin responsive DRG neurons both in RTX
pretreated laminectomy group (laminectomy 71.75±6.69% vs RTX laminectomy
45.0±2.04%, 4 rats each group,**p<0.01, Fig. 10) and sham rats(sham 59.51±2.82% vs
RTX and sham 33.60±1.50%, n=10 vs 5 rats each group, *P<0.001, Fig. 10).
Thus, on POD7, RTX still attenuated the laminectomy enhanced DRG neurons’
responses to capsaicin.
Effects of RTX on wheel running and home cage activity
We observed the whole week after laminectomy using RTX. And found significant
changes after RTX administration. The attenuated behavior was recovered faster
compared with laminectomy alone. Including running distance and locomotion.
rtx la m i D is ta n c e
B
a
s
e
lin
e
0
1
2
3
4
5
6
7
0
5 0
1 0 0
1 5 0
2 0 0
R T x + L a m in e cto m y lia n g
N o D ru g (N o A lz e t) L a m i lia n g
S h a m lia n g
rtx la m i M a x S p e e d
B
a
s
e
lin
e
0
1
2
3
4
5
6
7
0
5 0
1 0 0
1 5 0
R T x + L a m in e cto m y
L a m i
S h am
wo	
  	
  
On	
  pod6	
  7	
  there	
  is	
  a	
  sig	
  diff	
  between	
  RTX	
  and	
  no	
  drug	
  laminectomy	
  in	
  running	
  distance	
  (two	
  way	
  anova,	
  
for	
  multiple	
  t-­‐	
  test,	
  day	
  3,4,5,6,7	
  is	
  different.	
  P=0.016,	
  0.0013,0.0013,0.0002,0.0004).	
  On	
  pod0,	
  there	
  is	
  a	
  
diff	
  between	
  RTX	
  and	
  no	
  drug	
  in	
  maximum	
  speed(two	
  way	
  anova,multiple	
  t	
  test,	
  day	
  0,1,2,	
  
p=0.0003,0.01,0.02).	
  there	
  is	
  no	
  diff	
  between	
  the	
  2	
  group	
  in	
  acceleration.	
  
R T X F in e m o to r m o v e m e n t
B
a
s
e
lin
e
0
1
2
3
4
5
6
7
0
5 0
1 0 0
1 5 0
S h am
L a m in e c to m y
L a m in e cto m y -R T x
R T X R e a rin g
B
a
s
e
lin
e
0
1
2
3
4
5
6
7
0
5 0
1 0 0
1 5 0
S h a m lia n g
L a m in e c to m y
L a m in e cto m y R T x
there is a sig diff between RTX and no drug laminectomy in fine motor movement (two
way anova, for multiple t- test, day 1,2 is different. P=0.014, 0.026). there is a diff
between RTX and no drug in rearing(two way anova,multiple t test, day 1, p=0.008).
there is no diff between the 2 group in acceleration.
Effects of RTX on the thermal hyperalgesia
The effects of the intrathecal administration of RTX (200ng) on thermal hyperalgesia in
naïve rats at 7 days and 10 days after injection of RTX were investigated.
Our results show that the intrathecal administration of 200ng RTX inhibited the thermal
response of hindpaw (Fig. 11), which confirmed the valid intrathecal injection of RTX.
Discussion
The pain after operation represents a significant clinical challenge for treatment. The
search for new treatment options of postoperative pain has been directed toward
specific mediators and modulators of pain and inflammation.
In this study, we showed that laminectomy sensitized the DRG sensory neurons
indicated by elevated amplitude of calcium response curve and prolonged duration both
in response to high K and capsaicin and a higher percentage of capsaicin responsive
,TRPV1 positive neurons.
The present study also showed that it RTX attenuated DRG responsiveness to
potassium and capsaicin evidenced by recovered amplitude, reversion of prolonged
duration to high K and capsaicin and reduced percentage of TRPv1 positive DRG
neurons.
Furthermore, on pod7 laminectomy still has prolonged the duration of intracellular
neuronal calcium response to capsaicin, which is also reversed by pretreatment of i.t.
RTX indicated by a normalized duration of calcium response. This showed an extended
effect of RTX.
Laminectomy produces a decrease in rat wheel running and spontaneous home cage
activities. I .T. RTX pretreatment reversed the decrease of wheel running distance and
recovered the rearing and fine movement. Because there were no sided effects
observed in the experiment, thus the safety profile of RTX was established. We propose
that RTX intrathecal application could be a useful strategy in alleviating postoperative
pain.
We developed an rat model of postoperative pain and examined whether TRPV1 play a
role in mediating postoperative pain injury. In sensory dorsal root ganglia, the trpv1 are
expressed on small and medium –sized neurons and are mainly involved with mediation
of nociceptive information. TRPV1 is expressed by the type II A-delta and C-fiber
neurons, functioning as a molecular integrator of nociception. TRPV1-positive neurons
are essential for the development of mechanical allodynia.
Trvp1 is sensitized by phosphorylation of trpv1 via cAMP8
. and we tested he effect of
specific TRPV1 agonists could overexcited DRG neurons and provide a potential way of
relieving pain.
Prior studies have shown that pain could induce expression of TRPV1 and neuron
phenotype change. Nerve injury and inflammation could enhance the expression of
trpv1 including the transcription, translation, post translational modification and
trafficking of trpv1(Hucho T 2007 neuron, zhang x, EMBO J 2005, Ji RR, Neuron 2002).
The net effect is central and peripheral sensitization. This could switch the phenotype of
DRG nruosns and spinal cord neurons from naïve state to an altered state. Thus
redcuce the pain threshold(Patapoutian A, 2009).Trpv1 could enhanced form gene
level, translational level (mRNA), and post-translational modifications such as
phosphorylation, n-glycosylation and covalent modification9
. Especially phosphorylation
by PKCᵋ (Cesare 1999, Mandadi S 2006 pain) 10
and PKA(Bhave 2002, Lopshire, 1998).
Also there is evidence showing the phosphorylation of Trpv1 is enhanced. .Inflammaorty
mediators sensitized trpv1 through phosphorylation of trpv1 by protein kinase PKCᵋ
(Mandadi S 2006 Pain zhang H, 2007 J neuroscience).
In postoperative rat, trpv1 could be elevated because of gene level, rna level and
posttranstlational level change. Recent study demonstrated that TRpv1 antagonist
intratheally and local application both can allevievate thermal hyperalgsiea and
mechanical allodynia(Uchytilova mole pain ,2014 pure behavior). Also in a model of
thoracotomy postoperative pain, RTX significantly reduced mechanical allodynia
following surgery.(Shin Jw , 2010). TRPV1-expressing C-fibers are sensitized to heat
and acid after incision (Kang S 2010 Pain), All this indicated the involvement of trpv 1 in
postoperative pain. As we have demonstrated here, there were marked differences in
small DRG neurons between laminectomy group and sham group with respect to the
magnitude and decay of capsaicin-evoked Calcium transients. In order to identify
neurons innervating the site of surgery, DiI was injected in lumbar region prior to the
surgery. One day after surgery, there was a significant increase in the magnitude and
decrease in the decay of capsaicin evoked calcium transients in small DRG neurons.
Similar decreases in decay were also observed in the transients evoked by capsaicin
even 7 days after laminectomy. The reason of the enhanced intracellular calcium
transients during inflammation could be the inflammatory mediators such as nerve
growth factor because they might affect the calcium efflux, uptake and binding of
calcium and thus contribute to the enhanced calcium transients during inflammation (Lu,
2008 ). The trpv1 sensitaztion in laminectomy group could be attributed to inflammatory
mediators such as Nerve growth factor (NGF) because it induces sensitization of the
TRPV1(Zhu W, 2007 mol cell neurosci). Other Inflammatory mediators (BK, substance
P) also modulate TRPV1 activity (Sugiura T. Bradykinin lowers the threshold
temperature for heat activation of vanilloid receptor
1. J Neurophysiol 2002; 88:544–8. Neurokinin-1 receptor enhances TRPV1 activity in
primary sensory neurons via PKCepsilon: a novel pathway for heat hyperalgesia. J
Neurosci. 2007 Zhang H). Studies have shown that trpv1 sensitization(by heat)
produced a larger membrane deploarizations in rat DRGs. This could lower the
threshold and increase excitability of DRG neurons. (Neelands, 2008 inflam research,
zhang xf, 2011). We demonstrated that surgery induced an enhanced calcium response
to both potassium and capsaicin. Because Intracellular calcium plays a critical role in
mediation of many cellular processes such as transcription, ion channel activity, it
regulates the neuronal excitability and transmitter release (Berridge, 2005)and hormone
release, and Ca2 +
-dependent gene transcription. , so the enhanced calcium response
seen in small DRG after surgery might contribute to the postoperative pain by changing
afferent excitability and facilitating transmitter transcription and release from the
nociceptive afferents. Indeed, pervious study showed that activation of trpv1 in afferent
terminals could increase the rate of glutamate release by allowing calcium into neurons
(Shoudai 2010). Such results suggest that activated, presynaptic TRPV1+ receptors
trigger continuous resting release of glutamate vesicles at physiological temperatures
only in capsaicin-responsive terminals. Thus the sensitization of DRGs found in
postoperative pain model indicated the higher excitability of DRG neurons and
increased neurotransmitter release in afferent terminals.
In terms of percentages of trpv1 response positive cells, unregulated trpv1
responsiveness in primary-afferent fibers occurs in inflammatory disease and irritable
bowel syndrome (yiangou 2001, Chan 2003). Perivous study has shown that nerve
injure leads to immnunoreactivity changes from trpv1 negative to positive in large
diameter neurons after nerve injury(Hudson 2001). A similar gain of capsaicin sensitivity
was seen in calcium imaging recently after nerve injury (Kim 2014 neuron). These
phenotypic switch was mostly seen in nerve injury and chronic pain(Costigan M 2009
Annu Rev Neurosci). Interestingly, cancer pain also leads to increase of trpv1
expression in both myelinated DRG neurons and perptidergic unmyelinated DRG
neurons (Niiyama Y, 2007 neuroscience). In our experiment, the intracellular calcium
response to capsaicin is also enhanced in terms of percentages of positive cells. The
percentage of TRPV1 responsive DRG rose in pain group was higher compared with
control group. The percentage change might reflect a phenotype change in DRG
population. This findings extended previous research results by showing that
unregulated trpv1 responsiveness in primary-afferent fibers occurs in postoperative
pain,
Together, these data are consistent with the hypothesis that the TRPV1 contribute to
postoperative pain DRG excitability. Indeed, the responses are both elevated in pod1
and pod7 when stimulated with capsaicin. To further understand the role to trpv1-
positive neurons in development of postoperative pain, we studied the Pod7 response in
DRGS. On PoD7, We found the the amplitude is reduced and d the percentage is
recovered. The amplitudes of capsaicin evoked calcium transients and percentages of
capsaicin responsive neurons are not higher in surgery group when compared with
control. This showed that the surgery induced a recoverable enhanced response in
DRGS indicating the disappearance of the inflammaotory part of postoperative pain.
However on pod7 the duration of capsaicin induced calcium response remains longer
in surgery group. This could due to the long- lasting remaining sensitization of DRGS
after surgery. This extended effect of surgery on calcium responses was reflected by
our wheel running results, which showed that on Pod7 there is still a difference between
RTX and no treatment group in running distance(On pod6 and pod7 there is a sig diff
between RTX and no drug laminectomy in running distance (two way anova, for multiple
t- test, day 3,4,5,6,7 is different. P=0.016, 0.0013,0.0013,0.0002,0.0004).
In our study, One day after the laminectomy, there was also a significant increase in the
magnitude and decrease in the decay of potassium evoked calcium transients in DiI
labeled neurons. We found the response duration, amplitude of DRG response to k30
to be elevated on POD1. And this enhanced response disappeared on POD7. Evidence
showed that the nerve injury in DRG leads to decrease in resting calcium concentration
(Fuchs ,2005) and a decrease in both the magnitude and decay of evoked calcium
transients (Fuchs 2007)and nerve injury also leads to a decreased voltage gated
calcium currents in injured DRG (MaCallum 2003, Baccei and Kocsis,2000). Also there
was reports that inflammation is associated with increases in magnitude and decrease
in decay of the high potassium evoked transient in small putative nociceptive
afferents(Lu 2008,neuroscience). In the present postoperative pain model, high
potassium induced weak excitation of naïve DRG , but strongly activate the DRG in
laminectomy group as indicated by increase in calcium transients’ amplitudes and
durations. This augmented responsiveness indicated neuronal sensitization. This
stronger activation is possibly due to trpv1 sensitization in laminectomy group because
inflammatory mediators induces an acute sensitization of nociceptive DRG neurons
partially through sensitization of the TRPV1(Zhu W, 2007 mol cell neurosci). The high
responsiveness to KCl and capsaicin shown in our postoperative pain model support
the idea of sensitization of DRG and phenotypic changes in DRGs after surgery. This
enhanced responsiveness to potassium disappeared on POD7.
Using this pain model, we also showed that the significantly augmented DRG neurons’
response to both potassium and capsaicin challenges are blunted by the application of
TPV1 agonist RTX. Also the percentage changes happened following the surgery are
also prevented by pretreatment of RTX. RTX mediated antinociceptive effects both in
home cage locomotion and wheel running. Trpv1 agonists-induced analgesia is
proposed be due to receptor desensitization because of a sustained activation of
receptors()Jeffry Jap los one 2009, Raisinghani M 2005 j physiol). I. t. RTX couls cause
aa sustained increased freuqencey of spontaneous EPSCs and lead to a
desensitization response and depolarization block, synaptic failures(Jeffry Ja plosone
2009 sikand p, 2007 j physiol).. RTX has been shown to cause both spinal cord and
DRG changes if applied intrathecal.
RTX could induce a degenaeraion of trpv1 positive neurons by causing a neurotoxic
influx of calcium ion through the channels resulting a calcium induced neuronal
death(karai 2004). Our data extended the previous findings by showing that the
percentage of DRG neurons responveis to capsicien is significantly reduced aftet RTX
treatment. Also it has been shown that the ablation of central terminals of trv1
expressing neurons occured. after i.t. RTX (Bishinoi M 2011 j pain). Even when used
perineurally and remote from the neuronal perikary in DRG , RTX still could ablate DRG
TRPV1 positive neurons (Neubert 2008). RTX is neuroexcitotoxic and ablate neurons.
RTX is a promising drug for treatment because of its desensitizing effect and ablate
effect. Single dose might desensitize ,repeated dose or high dose could cause
neurotoxity and permanently degenerate nerve Trpvv1 + nerve endings (Ladarola
2011). RTX induced a prolonged increase in intracellular calcium in Trvp1 positive
neurons in human dorsal root ganglion cultures, , while leaving other adjacent neurons
unaffected(Karai, 2004). It has been proved that intrathecal RTX could reduce TRpv1,
CGRP and trpa1 immunoreactivities in the lumbar dorsal root ganglia and (karai 2004,
Neubert 2008 Klafke 2012 neuroscience), indicating the deltetion ot DRG TRPV1-
expresssing neurons(Neubert). Intrathecal rtx also produce ablation of central nerve
terminals of DRG neurons indicated by decreased immune reaction of TRV1 in spinal
cord dorsal horn (Bishnoi 2011). This cellular changes were reflectd in behavioral tests
such as prolonged hind paw thermal stimulation latency and carrageenan-induced
inflammatory therma lanalgesia(Bishnoi 2011). Our thermal test on hind paw confirmed
the prolonged radiant noxiouls thermal stimulus latency and suggested the specific
deletion of trpv1 positive neurons. In the present study, intrathecal RTX reduced the
calcium response. We found that RTX producing a significantly reduced responsiveness
to potassium and capsacin in DRGs.Also In calcium imaging, the percentages of trpv1
responsive DRG neurons decrease after RTX application both in postoperative rats and
sham rats further proved that the intrathecal RTX targeted trpv1 positive neurons in
DRG and made them inert to high potassium and capsaicin. We propose that i.t. RTX
also affect DRG trpv1 channels because RTX diffused to DRG and act on primary
afferent neurons. This may partly explained our finding that after rtx, the DRG response
to capsaicin are reduced.
This further extended the findings that RTX decreasing the TRPV1 immunoreactivities
in DRG after both perineural and intrathecal RTX application(neubert Jk, 2008, Klafke
2012 neuroscience)
RTX has been used for debilitating arthritic and cancer pain . In rats of postoperative
pain, RTX can significantly reduce the pain(my paper). Also the RTX has been shown to
be effective in treatment of neuropathic pain(gariel Tender, spinaljournal 2008) because
RTX jinjection acuse an excitotoxic effect when injected into sensory ganglia. RTX is a
transient receptor potential vanilloid 1 channel agonist. It also contributes to
inflammatory and heat allodynia relieve. Our intrathecal injection of RTX also cause an
inert DRG neuron reaction to K30 and capsaicin, both on pod1 and pod7. Extensive
research dta have shown that trpV1 positive neurons mediate thermal hyperalgesia in
pain models.(11-16 of Tender 2008 spine ournal). Moreover , the same cells apppeart
to be responsible ofre the mediation of neurogenic inflamesion(13.17-19), blockage of
trpv1 apears eefective in reversing established hyperalgesia associate with
inflammation(18). Our data corroborated that RTX could abolish the heat induce pain in
rats, evidence by the extenede time response time to the heat application on hindpaw in
rats treated with i.t. RTX. The paw became less responsive to heat. These results are in
concordance with previous studies showing theat trpv1-posticitve neurons mediated
termal hyperalgesia.
Also it is shown that inhibition of trpv1 centrally could inhibit the mechanical
hyperalgesia even it is not a mechanotransducer(Kim 2014 Neuron). So far there have
been no data to demonstrate the involvement of trpv1-positive neurons in mechanical
sensitivity or allodynia. However, some earlier reports showed that RTX injected in
DRGs could elevated the average withdrawal threshold of the paws and the magnitude
of touch –evoked allodynia was improved, which means the RTX improved tactile
allodynia via selectively eliminating the trv1-positive cells in the DRGs. This could be
explained by the cross-talk between A-beta, A-delta and C-fiber neurons in DRGs (23 of
tender 2008) . After operation, large amounts of glutamate and aspartate are released
into extracellular space and the inhibitory neurotransmitter GABA is reduced in spinal
dorsal horn (22) and also in the DRGs, resulting the hypersensitivity of A fibers and C
fibers. When use a touch test of allodynia, the A-beta fibers carried the signals to the
DRG and excited not only the A-beta cell bodies but also A-delta and C neurons
because of the cross-excitation. The excitation of a A-delta and C neurons subsequently
contribute to the touch induced pain and personal’s experience and emotional status.
Thus, deletion of trpv1 neurons (include A-delta and C fibers) could reduce the
excitation of A-delta and C- fibers in DRGs and thus reduce the whole animal’s
response to touch.
Also even we did not test the mechanical threshold of back region. There was evidence
showing that the surgery affected the cutaneous sensitivity in the back region after
thoracotomy and RTX could relieve the allodynia developed after(Shin 2010) surgery11
.
It can relieve pain like shown in behavioral tests wheel running and spontaneous
activities. It already has been shown when used in large doses intrathecally(3.8ug/kg), it
is able to alleviate capsaicin induced nocifensive behavior(bishnoi 2011 j pain) and both
capsaicin and carrageenan induced inflame thermal hypersensitivity	
  (bishnoi 2011 j
pain).	
  	
  Also it is shown that inhibition of trpv1 centrally could inhibit the mechanical
hyperalgesia even it is not a mechanotransducer(Kim 2014 Neuron).
We observed the spontaneous activity after surgery and the wheel running behavior
after laminectomy instead, it is shown that the wheel running and home cage activity are
changed by laminectomy and recovered partially due to RTX. We say the RTX
improved the running distance and the spontaneous activity in postoperative rats. Such
as rearing and fine movements in homecage, which proved that RTX mediated
antinociceptive effects and even after intrathecal RTX, animal maintain the motor
control. There was report that RTX caused a long lasting analgesia in cancer pain
model even up to 14 week after application and the hind paw heat response was
significantly reduced (Brown 2005). Our heat test further confirmed that intrathecal RTX
induced a higher heat response threshold. Also it has been shown that lumbar epidural
application of RTX resulted a focused distribution to lumbar neurons and lead to a
profound and segmental analgesia12
. Recently it was shown that intrathecal RTX could
lead to dose-dependent deletion of TRPV1 CGRP and phosphor ERK
immunoreactivities in superficial spinal cord such as laminae I and II. Other studies
showed the physiological sensation could recover 2 weeks after RTx treatment2
.
In summary, the high responsiveness to capsaicin and potassium in DRG reflected the
sensitization of neurons and this indicates high excitability and more neurotransmitter
release in peripheral nociceptive neurons. RTX is a promising pain killer because both
the calicium imaging and behavioral test support its use in postoperative pain.
1.	
   Mishra	
  SK,	
  Hoon	
  MA:	
  Ablation	
  of	
  TrpV1	
  neurons	
  reveals	
  their	
  selective	
  role	
  in	
  thermal	
  pain	
  
sensation.	
  Mol	
  Cell	
  Neurosci	
  2010;	
  43:	
  157-­‐63	
  
2.	
   Neubert	
  JK,	
  Mannes	
  AJ,	
  Karai	
  LJ,	
  Jenkins	
  AC,	
  Zawatski	
  L,	
  Abu-­‐Asab	
  M,	
  Iadarola	
  MJ:	
  Perineural	
  
resiniferatoxin	
  selectively	
  inhibits	
  inflammatory	
  hyperalgesia.	
  Mol	
  Pain	
  2008;	
  4:	
  3	
  
3.	
   Karai	
  L,	
  Brown	
  DC,	
  Mannes	
  AJ,	
  Connelly	
  ST,	
  Brown	
  J,	
  Gandal	
  M,	
  Wellisch	
  OM,	
  Neubert	
  JK,	
  Olah	
  Z,	
  
Iadarola	
  MJ:	
  Deletion	
  of	
  vanilloid	
  receptor	
  1-­‐expressing	
  primary	
  afferent	
  neurons	
  for	
  pain	
  control.	
  J	
  Clin	
  
Invest	
  2004;	
  113:	
  1344-­‐52	
  
4.	
   Zimmermann	
  M:	
  Ethical	
  guidelines	
  for	
  investigations	
  of	
  experimental	
  pain	
  in	
  conscious	
  animals.	
  
Pain	
  1983;	
  16:	
  109-­‐10	
  
5.	
   Lu	
  SG,	
  Zhang	
  X,	
  Gold	
  MS:	
  Intracellular	
  calcium	
  regulation	
  among	
  subpopulations	
  of	
  rat	
  dorsal	
  
root	
  ganglion	
  neurons.	
  J	
  Physiol	
  2006;	
  577:	
  169-­‐90	
  
6.	
   Waxman	
  SG,	
  Dib-­‐Hajj	
  S,	
  Cummins	
  TR,	
  Black	
  JA:	
  Sodium	
  channels	
  and	
  pain.	
  Proc	
  Natl	
  Acad	
  Sci	
  U	
  S	
  
A	
  1999;	
  96:	
  7635-­‐9	
  
7.	
   Takahashi	
  Y,	
  Chiba	
  T,	
  Kurokawa	
  M,	
  Aoki	
  Y:	
  Dermatomes	
  and	
  the	
  central	
  organization	
  of	
  
dermatomes	
  and	
  body	
  surface	
  regions	
  in	
  the	
  spinal	
  cord	
  dorsal	
  horn	
  in	
  rats.	
  J	
  Comp	
  Neurol	
  2003;	
  462:	
  
29-­‐41	
  
8.	
   Bhave	
  G,	
  Zhu	
  W,	
  Wang	
  H,	
  Brasier	
  DJ,	
  Oxford	
  GS,	
  Gereau	
  RWt:	
  cAMP-­‐dependent	
  protein	
  kinase	
  
regulates	
  desensitization	
  of	
  the	
  capsaicin	
  receptor	
  (VR1)	
  by	
  direct	
  phosphorylation.	
  Neuron	
  2002;	
  35:	
  
721-­‐31	
  
9.	
   Voolstra	
  O,	
  Huber	
  A:	
  Post-­‐Translational	
  Modifications	
  of	
  TRP	
  Channels.	
  Cells	
  2014;	
  3:	
  258-­‐87	
  
10.	
   Cesare	
  P,	
  Dekker	
  LV,	
  Sardini	
  A,	
  Parker	
  PJ,	
  McNaughton	
  PA:	
  Specific	
  involvement	
  of	
  PKC-­‐epsilon	
  
in	
  sensitization	
  of	
  the	
  neuronal	
  response	
  to	
  painful	
  heat.	
  Neuron	
  1999;	
  23:	
  617-­‐24	
  
11.	
   Shin	
  JW,	
  Pancaro	
  C,	
  Wang	
  CF,	
  Gerner	
  P:	
  The	
  effects	
  of	
  resiniferatoxin	
  in	
  an	
  experimental	
  rat	
  
thoracotomy	
  model.	
  Anesth	
  Analg	
  2010;	
  110:	
  228-­‐32	
  
12.	
   Szabo	
  T,	
  Olah	
  Z,	
  Iadarola	
  MJ,	
  Blumberg	
  PM:	
  Epidural	
  resiniferatoxin	
  induced	
  prolonged	
  regional	
  
analgesia	
  to	
  pain.	
  Brain	
  Res	
  1999;	
  840:	
  92-­‐8	
  

More Related Content

What's hot

Dissertation (rev 2011-05-23)_final_pdf
Dissertation (rev 2011-05-23)_final_pdfDissertation (rev 2011-05-23)_final_pdf
Dissertation (rev 2011-05-23)_final_pdfWalter Babiec
 
CHM462 Poster Presentation By Alexander Ward (1)
CHM462 Poster Presentation By Alexander Ward (1)CHM462 Poster Presentation By Alexander Ward (1)
CHM462 Poster Presentation By Alexander Ward (1)Alexander Ward
 
SfN 2008_Tabata_CruzAguado_Progranulin
SfN 2008_Tabata_CruzAguado_ProgranulinSfN 2008_Tabata_CruzAguado_Progranulin
SfN 2008_Tabata_CruzAguado_ProgranulinPierre Zwiegers
 
Pancetti et al 2004 methilene
Pancetti et al 2004 methilenePancetti et al 2004 methilene
Pancetti et al 2004 methileneJorge Parodi
 
RESVERATROL DERIVATIVES WITH INCREASED BIOAVAILABILITY
RESVERATROL DERIVATIVES WITH INCREASED BIOAVAILABILITYRESVERATROL DERIVATIVES WITH INCREASED BIOAVAILABILITY
RESVERATROL DERIVATIVES WITH INCREASED BIOAVAILABILITYGanesan Venkatesan
 
Summer Research Poster
Summer Research PosterSummer Research Poster
Summer Research PosterAlan Kim
 
Replacement Polymorphism of DENV E Asn-67 with Lys-67
Replacement Polymorphism of DENV E Asn-67 with Lys-67Replacement Polymorphism of DENV E Asn-67 with Lys-67
Replacement Polymorphism of DENV E Asn-67 with Lys-67vivishout
 
Replacement Polymorphism of N-linked Glycosylation site Asn-67 in DENV E
Replacement Polymorphism of N-linked Glycosylation site Asn-67 in DENV EReplacement Polymorphism of N-linked Glycosylation site Asn-67 in DENV E
Replacement Polymorphism of N-linked Glycosylation site Asn-67 in DENV Evivishout
 
Mileyshmi Holder Viera-EB poster-April
Mileyshmi Holder Viera-EB poster-AprilMileyshmi Holder Viera-EB poster-April
Mileyshmi Holder Viera-EB poster-AprilMileyshmi Holder
 
Reversal of neurological defects in a mouse model of rett syndrome
Reversal of neurological defects in a mouse model of rett syndromeReversal of neurological defects in a mouse model of rett syndrome
Reversal of neurological defects in a mouse model of rett syndromeJosé Luis Moreno Garvayo
 
Brian Covello: Diabetes Research Presentation Semester 2
Brian Covello: Diabetes Research Presentation Semester 2Brian Covello: Diabetes Research Presentation Semester 2
Brian Covello: Diabetes Research Presentation Semester 2Brian Covello
 
Ronan Talty - RISE 2015 Poster
Ronan Talty - RISE 2015 PosterRonan Talty - RISE 2015 Poster
Ronan Talty - RISE 2015 PosterRonan Talty
 
Science-2007-Kolodkin-Gal-652-5
Science-2007-Kolodkin-Gal-652-5Science-2007-Kolodkin-Gal-652-5
Science-2007-Kolodkin-Gal-652-5Ilana Kolodkin
 

What's hot (19)

Dissertation (rev 2011-05-23)_final_pdf
Dissertation (rev 2011-05-23)_final_pdfDissertation (rev 2011-05-23)_final_pdf
Dissertation (rev 2011-05-23)_final_pdf
 
CHM462 Poster Presentation By Alexander Ward (1)
CHM462 Poster Presentation By Alexander Ward (1)CHM462 Poster Presentation By Alexander Ward (1)
CHM462 Poster Presentation By Alexander Ward (1)
 
SfN 2008_Tabata_CruzAguado_Progranulin
SfN 2008_Tabata_CruzAguado_ProgranulinSfN 2008_Tabata_CruzAguado_Progranulin
SfN 2008_Tabata_CruzAguado_Progranulin
 
Pancetti et al 2004 methilene
Pancetti et al 2004 methilenePancetti et al 2004 methilene
Pancetti et al 2004 methilene
 
My Pain 2015
My Pain 2015My Pain 2015
My Pain 2015
 
RESVERATROL DERIVATIVES WITH INCREASED BIOAVAILABILITY
RESVERATROL DERIVATIVES WITH INCREASED BIOAVAILABILITYRESVERATROL DERIVATIVES WITH INCREASED BIOAVAILABILITY
RESVERATROL DERIVATIVES WITH INCREASED BIOAVAILABILITY
 
Summer Research Poster
Summer Research PosterSummer Research Poster
Summer Research Poster
 
Replacement Polymorphism of DENV E Asn-67 with Lys-67
Replacement Polymorphism of DENV E Asn-67 with Lys-67Replacement Polymorphism of DENV E Asn-67 with Lys-67
Replacement Polymorphism of DENV E Asn-67 with Lys-67
 
Replacement Polymorphism of N-linked Glycosylation site Asn-67 in DENV E
Replacement Polymorphism of N-linked Glycosylation site Asn-67 in DENV EReplacement Polymorphism of N-linked Glycosylation site Asn-67 in DENV E
Replacement Polymorphism of N-linked Glycosylation site Asn-67 in DENV E
 
Mileyshmi Holder Viera-EB poster-April
Mileyshmi Holder Viera-EB poster-AprilMileyshmi Holder Viera-EB poster-April
Mileyshmi Holder Viera-EB poster-April
 
Reversal of neurological defects in a mouse model of rett syndrome
Reversal of neurological defects in a mouse model of rett syndromeReversal of neurological defects in a mouse model of rett syndrome
Reversal of neurological defects in a mouse model of rett syndrome
 
Brian Covello: Diabetes Research Presentation Semester 2
Brian Covello: Diabetes Research Presentation Semester 2Brian Covello: Diabetes Research Presentation Semester 2
Brian Covello: Diabetes Research Presentation Semester 2
 
Alewel_poster
Alewel_posterAlewel_poster
Alewel_poster
 
FOS ALS '13 Poster (final)
FOS ALS '13 Poster (final)FOS ALS '13 Poster (final)
FOS ALS '13 Poster (final)
 
My Researches
My ResearchesMy Researches
My Researches
 
Final Poster
Final PosterFinal Poster
Final Poster
 
Ronan Talty - RISE 2015 Poster
Ronan Talty - RISE 2015 PosterRonan Talty - RISE 2015 Poster
Ronan Talty - RISE 2015 Poster
 
Science-2007-Kolodkin-Gal-652-5
Science-2007-Kolodkin-Gal-652-5Science-2007-Kolodkin-Gal-652-5
Science-2007-Kolodkin-Gal-652-5
 
103.full
103.full103.full
103.full
 

Similar to Effects of laminectomy and TRPV1 modulation on calcium signaling and behavioral outcomes in DRG neurons

Molecular Mechanisms of Pain. Part 2
Molecular Mechanisms of Pain. Part 2Molecular Mechanisms of Pain. Part 2
Molecular Mechanisms of Pain. Part 2SSA KPI
 
Application of PEA reduces WDR RF size - Final edit
Application of PEA reduces WDR RF size - Final editApplication of PEA reduces WDR RF size - Final edit
Application of PEA reduces WDR RF size - Final editShaun Croft, MScR
 
ASEA > Endurance Performance Study
ASEA > Endurance Performance StudyASEA > Endurance Performance Study
ASEA > Endurance Performance StudyASEA
 
Epidural Spinal Cord Stimulation for Modulating Small Sensory Nerve Fibers to...
Epidural Spinal Cord Stimulation for Modulating Small Sensory Nerve Fibers to...Epidural Spinal Cord Stimulation for Modulating Small Sensory Nerve Fibers to...
Epidural Spinal Cord Stimulation for Modulating Small Sensory Nerve Fibers to...stimwave
 
Stellar-Telemetry-Hypertension-Poster
Stellar-Telemetry-Hypertension-PosterStellar-Telemetry-Hypertension-Poster
Stellar-Telemetry-Hypertension-PosterTracie Rindfield
 
Effect of Biofield Treatment on Spectral Properties of Paracetamol and Piroxicam
Effect of Biofield Treatment on Spectral Properties of Paracetamol and PiroxicamEffect of Biofield Treatment on Spectral Properties of Paracetamol and Piroxicam
Effect of Biofield Treatment on Spectral Properties of Paracetamol and Piroxicamalbertdivis
 
Effect of Biofield Treatment on Spectral Properties of Paracetamol and Piroxicam
Effect of Biofield Treatment on Spectral Properties of Paracetamol and PiroxicamEffect of Biofield Treatment on Spectral Properties of Paracetamol and Piroxicam
Effect of Biofield Treatment on Spectral Properties of Paracetamol and PiroxicamMahendra Kumar Trivedi
 
Comparison of the Hypocalcemic Effects of Erythropoietin and U-74389G
Comparison of the Hypocalcemic Effects of Erythropoietin and U-74389GComparison of the Hypocalcemic Effects of Erythropoietin and U-74389G
Comparison of the Hypocalcemic Effects of Erythropoietin and U-74389Gasclepiuspdfs
 
Primer-directed enzymatic amplification of DNA with a thermostable DNA polyme...
Primer-directed enzymatic amplification of DNA with a thermostable DNA polyme...Primer-directed enzymatic amplification of DNA with a thermostable DNA polyme...
Primer-directed enzymatic amplification of DNA with a thermostable DNA polyme...José Luis Moreno Garvayo
 
Dr. Robert Langer - Simposio Internacional 'Terapias oncológicas avanzadas'
Dr. Robert Langer - Simposio Internacional 'Terapias oncológicas avanzadas'Dr. Robert Langer - Simposio Internacional 'Terapias oncológicas avanzadas'
Dr. Robert Langer - Simposio Internacional 'Terapias oncológicas avanzadas'Fundación Ramón Areces
 
PARP-1 Inhibitors In Oncology
PARP-1 Inhibitors In OncologyPARP-1 Inhibitors In Oncology
PARP-1 Inhibitors In OncologyGregory J. Wells
 
A Mathematical Bivariate Generalized Poisson Model for Cortisol Awakening Res...
A Mathematical Bivariate Generalized Poisson Model for Cortisol Awakening Res...A Mathematical Bivariate Generalized Poisson Model for Cortisol Awakening Res...
A Mathematical Bivariate Generalized Poisson Model for Cortisol Awakening Res...irjes
 
Sugarcane Ash and Sugarcane Ash-Derived Silica Nanoparticles Alter Cellular M...
Sugarcane Ash and Sugarcane Ash-Derived Silica Nanoparticles Alter Cellular M...Sugarcane Ash and Sugarcane Ash-Derived Silica Nanoparticles Alter Cellular M...
Sugarcane Ash and Sugarcane Ash-Derived Silica Nanoparticles Alter Cellular M...Arthur Stem
 
Using the Thermo Scientific Sorvall® WX Ultracentrifuge to Isolate Skeletal M...
Using the Thermo Scientific Sorvall® WX Ultracentrifuge to Isolate Skeletal M...Using the Thermo Scientific Sorvall® WX Ultracentrifuge to Isolate Skeletal M...
Using the Thermo Scientific Sorvall® WX Ultracentrifuge to Isolate Skeletal M...Daniel Schroen, PhD
 

Similar to Effects of laminectomy and TRPV1 modulation on calcium signaling and behavioral outcomes in DRG neurons (20)

Molecular Mechanisms of Pain. Part 2
Molecular Mechanisms of Pain. Part 2Molecular Mechanisms of Pain. Part 2
Molecular Mechanisms of Pain. Part 2
 
Application of PEA reduces WDR RF size - Final edit
Application of PEA reduces WDR RF size - Final editApplication of PEA reduces WDR RF size - Final edit
Application of PEA reduces WDR RF size - Final edit
 
ASEA > Endurance Performance Study
ASEA > Endurance Performance StudyASEA > Endurance Performance Study
ASEA > Endurance Performance Study
 
Epidural Spinal Cord Stimulation for Modulating Small Sensory Nerve Fibers to...
Epidural Spinal Cord Stimulation for Modulating Small Sensory Nerve Fibers to...Epidural Spinal Cord Stimulation for Modulating Small Sensory Nerve Fibers to...
Epidural Spinal Cord Stimulation for Modulating Small Sensory Nerve Fibers to...
 
VGCC
VGCCVGCC
VGCC
 
Stellar-Telemetry-Hypertension-Poster
Stellar-Telemetry-Hypertension-PosterStellar-Telemetry-Hypertension-Poster
Stellar-Telemetry-Hypertension-Poster
 
Research Article Presentation.pptx
Research Article Presentation.pptxResearch Article Presentation.pptx
Research Article Presentation.pptx
 
BATA2016 poster_official
BATA2016 poster_officialBATA2016 poster_official
BATA2016 poster_official
 
Effect of Biofield Treatment on Spectral Properties of Paracetamol and Piroxicam
Effect of Biofield Treatment on Spectral Properties of Paracetamol and PiroxicamEffect of Biofield Treatment on Spectral Properties of Paracetamol and Piroxicam
Effect of Biofield Treatment on Spectral Properties of Paracetamol and Piroxicam
 
Effect of Biofield Treatment on Spectral Properties of Paracetamol and Piroxicam
Effect of Biofield Treatment on Spectral Properties of Paracetamol and PiroxicamEffect of Biofield Treatment on Spectral Properties of Paracetamol and Piroxicam
Effect of Biofield Treatment on Spectral Properties of Paracetamol and Piroxicam
 
Comparison of the Hypocalcemic Effects of Erythropoietin and U-74389G
Comparison of the Hypocalcemic Effects of Erythropoietin and U-74389GComparison of the Hypocalcemic Effects of Erythropoietin and U-74389G
Comparison of the Hypocalcemic Effects of Erythropoietin and U-74389G
 
Primer-directed enzymatic amplification of DNA with a thermostable DNA polyme...
Primer-directed enzymatic amplification of DNA with a thermostable DNA polyme...Primer-directed enzymatic amplification of DNA with a thermostable DNA polyme...
Primer-directed enzymatic amplification of DNA with a thermostable DNA polyme...
 
Dr. Robert Langer - Simposio Internacional 'Terapias oncológicas avanzadas'
Dr. Robert Langer - Simposio Internacional 'Terapias oncológicas avanzadas'Dr. Robert Langer - Simposio Internacional 'Terapias oncológicas avanzadas'
Dr. Robert Langer - Simposio Internacional 'Terapias oncológicas avanzadas'
 
PARP-1 Inhibitors In Oncology
PARP-1 Inhibitors In OncologyPARP-1 Inhibitors In Oncology
PARP-1 Inhibitors In Oncology
 
ACCP publication
ACCP publication ACCP publication
ACCP publication
 
GLP-1 Poster_02JUN15
GLP-1 Poster_02JUN15GLP-1 Poster_02JUN15
GLP-1 Poster_02JUN15
 
A Mathematical Bivariate Generalized Poisson Model for Cortisol Awakening Res...
A Mathematical Bivariate Generalized Poisson Model for Cortisol Awakening Res...A Mathematical Bivariate Generalized Poisson Model for Cortisol Awakening Res...
A Mathematical Bivariate Generalized Poisson Model for Cortisol Awakening Res...
 
Sugarcane Ash and Sugarcane Ash-Derived Silica Nanoparticles Alter Cellular M...
Sugarcane Ash and Sugarcane Ash-Derived Silica Nanoparticles Alter Cellular M...Sugarcane Ash and Sugarcane Ash-Derived Silica Nanoparticles Alter Cellular M...
Sugarcane Ash and Sugarcane Ash-Derived Silica Nanoparticles Alter Cellular M...
 
Using the Thermo Scientific Sorvall® WX Ultracentrifuge to Isolate Skeletal M...
Using the Thermo Scientific Sorvall® WX Ultracentrifuge to Isolate Skeletal M...Using the Thermo Scientific Sorvall® WX Ultracentrifuge to Isolate Skeletal M...
Using the Thermo Scientific Sorvall® WX Ultracentrifuge to Isolate Skeletal M...
 
Visconti Poster
Visconti PosterVisconti Poster
Visconti Poster
 

Effects of laminectomy and TRPV1 modulation on calcium signaling and behavioral outcomes in DRG neurons

  • 1. A B   30μm   30μm   Figure  1  Representa5ve   epifluorescence  graphs  of  cultured  rat   DRG  neurons  before  (A)  and  aCer  (B)   applica5on  of  100nM  capsaicin.   Arrows,  Fura-­‐2-­‐loaded  cells  exhibi5ng   Ca2+    mobiliza5on  in  response  to   capsaicin.  (Scale  bars,  30μm.)  
  • 2. 0   0.5   1   1.5   2   2.5   3   3.5   4   0   150   300   450   600   750   900   1050   1200   (Sec)   0   0.5   1   1.5   2   2.5   0   150   300   450   600   (Sec)   ∆Ra5o  340/380  ∆Ra5o  340/380   Figure  2  Representa5ve  traces  of   capsaicin  evoked  calcium   responses  in  rat  DRG  neurons   (Blue),  enhanced  by  laminectomy   (Red).    Intrathecal  pretreatment   with  the  TRPV1  agonist  RTX   a^enuated  laminectomy  induced   enhancement  (Green).    
  • 3. Figure  3    On  POD1,  potassium  chloride  evoked  calcium  transients  in  rat  DRG   neurons  as  measured  by  the  ra5o  difference  between  the  peak  ra5o  and   baseline  ra5o  (∆Ra5o  340/380).  Data  are  presented  as  mean±SEM  of    ∆Ra5o   340/380.  ***p<0.001,  **p<0.01,  n=50-­‐80  cells  from  4-­‐6  rats  each  group.   Figure  4  On  POD1,  dura5on  (seconds)  of  recovery  to  80%  of  the  baseline  of  K+  evoked  Ca2+   transients  in  rats  DRG  neurons.  Data  are  presented  as  mean±SEM  of  the  5me  dura5on  to  80%   recovery  of  baseline  of  ∆Ra5o  340/380.  ***p<0.001,  **p<0.01,  n=  50-­‐80  cells  from  4-­‐6  rats  each   group.  
  • 4. Figure  5  On  POD1,  capsaicin  evoked  calcium  transients  in  rat  DRG  neurons  as   measured  by  the  ∆Ra5o  340/380.  Data  are  presented  as  mean±SEM  of    ∆Ra5o   340/380.  ***p<0.001,  *p<0.05,  n=45-­‐70  cells  from  4-­‐6  rats  each  group.   Figure  6  On  POD1,  dura5on  (seconds)  of  recovery  to  80%  of  the  baseline  of   capsaicin  evoked  Ca2+  transients  in  rats  DRG  neurons.  Data  are  presented  as   mean±SEM  of  the  5me  dura5on  to  80%  recovery  of  baseline  of  ∆Ra5o  340/380.   ***p<0.001,  *p<0.05,  n=  45-­‐70  cells  from  4-­‐6  rats  each  group.   Figure  7  On  POD1,  percentage  of  capsaicin  responsive  DRG  neurons.  Data  are   presented  as  mean±SEM  of  the  percentage  of  neurons  responsive  to  capsaicin  in   the  whole  DRG.  ***p<0.001,  *p<0.05,  n=  45-­‐70  cells  from  4-­‐6  rats  each  group.  
  • 5. Figure  8  On  POD7,  potassium  chloride  evoked  calcium  transients  in  rat  DRG   neurons  as  measured  by  the  ra5o  difference  between  the  peak  ra5o  and  baseline   ra5o  (∆Ra5o  340/380).  Data  are  presented  as  mean±SEM  of    ∆Ra5o  340/380.   **p<0.01,  n=45-­‐70  cells  from  4-­‐6  rats  each  group.    Figure  9  On  POD7,  dura5on  (seconds)  of  recovery  to  80%  of  the  baseline  of  K+   evoked  Ca2+  transients  in  rats  DRG  neurons.  Data  are  presented  as  mean±SEM  of   the  5me  dura5on  to  80%  recovery  of  baseline  of  ∆Ra5o  340/380.  **p<0.01,  n=   45-­‐70  cells  from  4-­‐6  rats  each  group.     la m in e c to m y s h a m s u rg e ry 0 .0 0 .5 1 .0 1 .5 DRatio340/380 la m in e c to m y s h a m s u rg e ry 0 5 0 1 0 0 1 5 0 t80%(seconds)
  • 6. Figure  10  On  POD7,  capsaicin  evoked  calcium  transients  in  rat  DRG  neurons  as   measured  by  the  ra5o  difference  between  the  peak  ra5o  and  baseline  ra5o   (∆Ra5o  340/380).  Data  are  presented  as  mean±SEM  of    ∆Ra5o  340/380.   ***p<0.001,  n=40-­‐60  cells  from  4-­‐6  rats  each  group.    Figure  11  On  POD7,  dura5on  (seconds)  of  recovery  to  80%  of  the  baseline  of  K+  evoked   Ca2+  transients  in  rats  DRG  neurons.  Data  are  presented  as  mean±SEM  of  the  5me   dura5on  to  80%  recovery  of  baseline  of  ∆Ra5o  340/380.  ***P<0.001,  **p<0.01,  n=  40-­‐60   cells  from  4-­‐6  rats  each  group.     Figure  12  On  POD7,  percentage  of  capsaicin  responsive  DRG  neurons.  Data  are   presented  as  mean±SEM  of  the  percentage  of  neurons  responsive  to  capsaicin  in  the   whole  DRG.  ***p<0.001,  n=  40-­‐60  cells  from  4-­‐6  rats  each  group.    
  • 7. On  pod6  7  there  is  a  sig  diff  between  RTX  and  no  drug  laminectomy  in  running   distance  (two  way  anova,  for  mul5ple  t-­‐  test,  day  3,4,5,6,7  is  different.  P=0.016,   0.0013,0.0013,0.0002,0.0004).  On  pod0,  there  is  a  diff  between  RTX  and  no  drug   in  maximum  speed(two  way  anova,mul5ple  t  test,  day  0,1,2,   p=0.0003,0.01,0.02).  there  is  no  diff  between  the  2  group  in  accelera5on.  
  • 8. there  is  a  sig  diff  between  RTX  and  no  drug  laminectomy  in  fine  motor   movement  (two  way  anova,  for  mul5ple  t-­‐  test,  day  1,2  is  different.  P=0.014,   0.026).  there  is  a  diff  between  RTX  and  no  drug  in  rearing(two  way   anova,mul5ple  t  test,  day  1,  p=0.008).  there  is  no  diff  between  the  2  group  in   accelera5on.  
  • 9. Figure  13  On  baseline,  post  RTX  injec5on  day  7,  post  RTX   injec5on  day  14,  thermal  pain  withdrawal  threshold  with  a   cutoff  of  25  seconds.  *p<0.05,  n=4  rats  each  group.  
  • 10. Prolonged and enhanced rodent intracellular Ca2+ signal in dorsal root ganglion after laminectomy was shortened and down regulated by intrathecal resiniferatoxin Abstract Background: postoperative pain is a challenging problem in clinic, compromising quality of life and leading to extended hospital stay and economic burden and could develop into chronic pain. Patients report significant pain and hypersensitivity postoperatively. This study examined the capsaicin and high potassium induced intracellular calcium response in cultured adult rat DRG neurons on postoperative day 1 and postoperative day 7, and examined the effects of resiniferatoxin treatment on neuronal intracellular calcium responses and rats behavior including wheel running and spontaneous home cage activities. Results: Calcium imaging showed that laminectomy resulted in prolonged duration and elevated amplitude of high potassium and capsaicin induced DRG neuron intracellular calcium responses. Also the surgery led to higher responsive ratio in DRG populations, compared to controls; this was reduced by intrathecal administration of 200ng resiniferatoxin. Laminectomy resulted in decreased wheel running distance, maximum speed , home cage rearing and fine movements. Resiniferatoxin partially reversed the effects of laminectomy on wheel running and home cage spontaneous activities. It also extended the heat response in hind paw after intrathecal injection. Conclusions: Laminectomu induces TRP sensitization and higher response to high potassium in DRG neurons and leads to reduced wheel running and spontaneous activities in home cage. These effects may be mitigated by preoperative treatment with
  • 11. TRPV1 agonist resiniferatoxin which could lead to desensitization of neurons, increase wheel running and alleviate spontaneous pain. Introduction A large body of evidence point to the importtant role of TrpV1, the capsaicin receptor, in sensation of noxious heat (Caterina et al., 1997). TrpV1is also believed to play a role in increased pain sensitivity following inflammation at least in part because inflammation induced phosphorylation of TrpV1 results in increased sensitivity of this ion channel to temperature (Bhave et al., 2002; Cesare et al., 1999; Nilius et al., 2005; Numazaki et al., 2002). Attention has focused on TrpV1 and TrpV1 cells for their role in pain sensation and as targets for pain therapies. Notably, TrpV1 antagonists have been shown to be efficacious in animal models of chronic pain(Szallasi et al.,2007;Wong andGavva,2009) but, as yet, have not been exploited in human pain management. An alternate strategy for pain management involves ablation of TrpV1-expressing cells or TrpV1 fibers. This approach has a long history: more than 25 years ago, capsaicin injection was reported to dramatically decrease the level of inflammatory mediators (e.g., substance P) and pain sensation for the life of animals (Jancso et al., 1977; Nagy et al., 1981; Yaksh et al., 1979). More recently, the potent TrpV1 agonist, resiniferotoxin (RTX) has been demonstrated to kill TrpV1-expressing neurons by affecting long-term Ca2+ entry (Olah eal., 2001). RTX has also been used therapeutically in animals and has been reported to alleviate chronic pain with no effects on discriminative touch or proprioception (Brown et al., 2005; Karai et al., 2004; Neubert et al., 2003). In a rat
  • 12. model, RTX was shown to eliminate many TrpV1 neurons, dramatically decrease responses to capsaicin and thermal pain (Karai et al., 2004), and affect mechanosensation (Tender et al., 2008). Mishra et al 1 carried out a detailed investigation into the efficacy of RTX in ablating sensory neurons and demonstrate that although a large subset of TrpV1neurons are killed by a single injection, other TrpV1 cells are not affected even by repeated RTX treatments. RTX reduces but does not eliminate capsaicin responses and thermal sensation of normal mice. We hypothesized that intrathecal application of RTX may block the DRB neurons temporarily and reduce the pain behavior. RTX is supposed to work on trpv1 positive small diameter neurons and leave intact the large diameter axons responsible for proprioceptive and somatic sensations.The intrathecal approach is proposed to broadly affect the DRGs surrounding the L4, L5 and their corresponding dermatome including the laminectomy site to mediate a selective antinociceptive effects. By modifying the trpv1 positive DRG neurons, this treatment can help relieve different clinical pain including the postoperative pain. Here, we set out to determine the function of TrpV1 cells and explored the use of RTX as a means to affect the intracellular calcium response to capsaicin and hight potassium stimulations. And to see if it can selectively eliminate trpv1 positive neurons in rats. The primary aim of this study was to determine whether incisional pain produced a higher calcium response in DRG neurons and how long it will last. The other aim of the study was to determine if intrathecal RTX will promote the reduction of calcium response in these DRG neurons after laminectomy and to check the effect of RTX on the hind paw
  • 13. thermal response and the effect of RTX on wheel running and rodent spontaneous pain behavior after intrathecal application. Low doses of RTX (100-250ng) has been proved to be effective when given perineurally2 , in single ganglionic injection or intrathecally3 . The minimus effective doses should be determined to avoid the po tential side effects considering the clinical settings. So this study used the 200ng RTX to block multiple ganglia by applying intrathecally. Materials and Methods Animals Experiments were conducted in adult male Sprague-Dawley rats (Harlan,Indianapolis, IN, USA) weighing 225-250 g. Animals were housed in groups of 2, maintained in a 12- hour light-dark cycle with a temperature-controlled environment and given food and water ad libitum. All procedures used in these experiments were reviewed and approved by the Institutional Animal Care and Use Committee at the University of Iowa. Animals were cared for and used in accordance with guidelines of the Committee for Research and Ethical Issues of the IASP4 . Chemicals and reagents 1,1'-Dioleyl-3,3,3',3'-Tetramethylindocarbocyanine methanesulfonate (DiI), Fura-2 acetoxymethyl (AM) ester and Pluronic F-127 (stock concentration 20%) were purchased from Invitrogen (Carlsbad, CA, USA). Concentrations were based on results from previous studies5 . DiI was dissolved in DMSO (25 mg/ml). Fura-2 AM (2.5µm) was dissolved in 1mM dimethyl sulfoxide and stored at -20°C. When ready to use, the Fura-2AM was diluted in bath solution (in mM: 130 NaCl, 5 KCl, 2 CaCl2, 1 MgCl2, 10
  • 14. Hepes, 10 glucose, and pH adjusted to 7.4, and osmolality adjusted with sucrose to 310-325 mOsm), together with Pluronic F-127 (0.025%) immediately before use. Resiniferatoxin (RTX), (Sigma Aldrich, St Louis, MO, USA) was dissolved in 10/10/80% mix of ethanol, Tween-80, and saline. Capsaicin (Sigma, St. Louis, MO, USA) was diluted to a final concentration of 1mM in 1.5% Tween-80, 1.5% alcohol and 97% saline. Capsaicin (100nM) was diluted in normal bath solution immediately before use. All other compounds were dissolved in normal bath solution directly. For obtaining high- potassium extracellular solution (K+ , 30mM), the K+ in normal bath solution was adjusted to 30 mM by iso-osmotic replacement of Na+ . Labeling DRG neurons Prior to tissue harvest (>5 days), sensory neurons innervating the lower back region were labelled with the retrograde tracer DiI. General anesthesia was induced with isoflurane (Abbott Laboratories, North Chicago, IL, USA) in room air and maintained at 1.5%, DiI solution (8 µl) was injected onto the right L4 and L5 laminae and surrounding muscles (4 µl ×2) by positioning the needle 2mm away from the midline (the L4 and L5 spinous processes) with a 30 g injection needle.     Intrathecal injection of RTX or Saline Under 1.5% isoflurane anesthesia, each rat in the RTX group received intrathecal (between the L4 and L5 vertebrae) injection of 20 µl RTX (200ng in 20 µl saline) using a Hamilton syringe and a 25-gauge needle. Each rat in saline group received injection of 20 µl saline.
  • 15. Behavioral pain tests Rats in the RTX group were tested on baseline, post injection day 7 and post injection day 14. For withdrawal latency to radiant heat stimuli, unrestrained rats were individually placed onto a 3-mm thick glass surface (40 × 40 cm) under a plastic chamber (21 × 27×15 cm) for approximately 15 minutes of acclimation. A focused radiant heat source was applied from underneath the glass floor onto the center of hind paw. The latency to evoke withdrawal was determined with a cut-off value of 30 seconds to prevent radiant heat evoked injury to the hindpaw. Each rat was tested 3 times, with a 5-minute interval between tests. The mean paw withdrawal latency was determined from 3 tests. Guarding pain was performed before withdrawal tests to avoid the potential influence of withdrawal responses on guarding pain. Wheel running and home cage locomotion tests Rats were placed individually in cages fitted with exercise wheels (Intellibio Activity Wheel, Seichamps, France). The rats were housed individually and allow to run on the exercise wheels with a computer that recorded the distance, speed, acceleration, revolutions and time (in one hour bins). A stable baseline was achieved after 14 days of running wheel exposure. The last 3 days were averaged to obtain the baseline. Data was recorded daily for 14 days. The distance traveled by the rat, acceleration, maximum speed, total motor movement, and rearing activity of rats became stable after 2 weeks
  • 16. habituation with this experimental protocol. The effects of a treatment on each rat were analyzed as percentage of the distance traveled during the baseline. Home Cage Activity Activity during the dark cycle (period when rats are active) after surgery was monitored. After acclimatization to individual home cages (17 x 5 x 18 inch) for 3 days, the cages were inserted into a SmartFrame Cage Rack Motor Monitor System (Kinder scientific, Poway, CA). Two grids of 7x15 infrared beams surrounded the cage; one was placed 6 cm above the floor to record total locomotor activity as total beam interruptions. The second series of beams was placed 18 cm above the cage floor and count rearing, when the rat stood on its hindpaws. Beam breaks were counted to record total activity and rearing. The data was then transmitted to a host computer and collected in 1-hour bins. The last 3 days were averaged as the baseline, and then rats would undergo sham surgery or full laminectomy. We used these monitors to examine hypermobility by acute pain stimuli. Laminectomy Procedure After the induction of general anesthesia with isoflurane in room air and maintained with 1.5% isoflurane. The surgery was performed with spontaneous ventilation through a nose cone. Each animal was shaved and sterilized with povidone iodine at the incision site. A 4cm midline longitudinal incision is made with a # 11 blade through the skin down to the fascia between L3-L6. Blunt dissection was continued through the incision into the muscle (removing muscle away from bone) to further divide and retracts the muscle. The posterior bony spine was exposed and L4-L6 spinous processes removed. The bone between L4-L5 was removed using a battery-powered drill. The dura was exposed without breaching it. After surgery, the wound was closed in layers; the muscle
  • 17. was closed with 4 sutures, followed by fascia using absorbable 3-0 vicryl suture (Ethicon, San Angelo, TX, USA). The skin was closed with 4 sutures of 3-0 nylon (Ethicon, San Angelo, TX, USA). Cell Culture Rats were euthanized with 100% carbon dioxide and the right-sided L1-L3 DRG were surgically removed and desheathed in ice-cold Hanks balanced salt solution (HBSS) (Gibco, Gaithersburg, MD, USA). DRGs were incubated 20 min at 37°C in 3 ml HBSS containing 0.125% papain (Worthington, Lakewood, NJ, USA) and then centrifuged at 800 g for 1 min. The papain solution was removed and replaced and incubated for 20 min with 3ml HBSS containing 10 mg collagenase IV (Worthington, Lakewood, NJ, USA) and 14mg dispase (Invitrogen, Carlsbad, CA, USA). DRGs were then centrifuged at ∼800 g for 1 min, resuspended with culture media F12 (Gibco, Gaithersburg, MD, USA). DRGs were mechanically dissociated in this solution with fire- polished Pasteur pipettes and then plated onto glass coverslips, previously coated by a solution of 0.1 mg/ml poly-D-lysine (Sigma Aldrich, St Louis, MO,USA) and 5 µg/ml mouse laminin (BD Bioscience, Bedford, CA, USA). A 0.5-mm-thick silicone ring with a 4-mm-diameter opening sealed to the surface of each well reduced the plating area in each well. The cells were incubated at 37°C, in 5% CO2 for 2 h. After 2 h, the wells were filled with 2ml F12 media (Gibco, Gaithersburg, MD, USA) containing 10%(v/v) Fetal Bovine Serum (FBS), 50 U/ml penicillin , 50 µg/ml streptomycin, and 50ng/ml nerve growth factor (NGF) (Sigma Aldrich, St Louis, MO,USA), and incubated overnight.
  • 18. Subclassification of DRG neurons Neurons were divided into small (<30 µm), and large (≥30 µm) based on cell body diameter as measure with a calibrated eyepiece reticule. Only neurons with visible nucleus are used. Neurons with a small cell body diameter are likely to be nociceptive afferents, while those with a large cell body diameter are likely to be non-nociceptive afferents6 . Intracellular Ca2+ measurements All experiments were performed within 24h of tissue harvest. DRG neurons were loaded with the calcium indicator dye, Fura-2 AM ester (2.5µM) along with 0.025% Pluronic for 20 min at room temperature as previously described 5 . Following fura-2 loading, DRG neurons were placed in a recording chamber mounted on the stage of an inverted IX-71 microscope (Olympus IX71, Japan) and were continuously perfused for 5 minutes with normal bath solution before the experiment began. Fluorescence data were acquired on computer running Slidebook 5.0 software (Molecular Devices, Sunnyvale, CA, USA) at 20°C. Only cells with DiI label were used for calcium imaging. Fluorescence was alternately excited at 340nm and 380nm using the Lambda DG-4 filter changer (Sutter Instruments, Novato, CA, USA) via a 10x (Numerical Aperture NA=0.30) or 20x (NA=0.70) objectives. Emitted fluorescence was collected at 510nm using an ORCA- 03G cooled digital CCD camera (Hamamatsu Photonics, Japan). Pairs of images were sampled at 1-3 Hz. For each field, an average of 20-40 cells for the 20x objective were tracked in real time. The ratio of fluorescence emission (510 nm) in response to 340/380 nm excitation was acquired at 1-3 Hz during drug applications. All drugs were applied
  • 19. via a gravity-fed chamber perfusion system. Cells were depolarized by capsaicin (100 nM, 30 seconds) or K+ (30 mM, 30 seconds). Cells not responsive to K+ were excluded. Two properties of calcium influx were measured: peak ratio (340/380 nm) of the Fura-2 signal, and duration of the Fura-2 signal (calculated as time to 80% of baseline value). In case of capsaicin depolarization, the percentages of cells responsive to capsaicin were also compared between groups. Statistical analysis Data are expressed as mean± standard error of the mean (SEM) unless otherwise stated. The “n” for each group refers to the number of neurons. Statistical significance was determined using Student’s t-test, or ANOVA when appropriate followed by Tukey’s post-hoc comparisons (GraphPad Prism software, San Diego, CA, USA). Differences at the P<0.05 level were considered statistically significant. Results To assess the effect of laminectomy on the DRG neurons’ intracellular calcium transients’ responses to K+ or capsaicin induced depolarization, Fura-2 Ca2+ imaging experiments were performed on DRG neurons that were transiently depolarized with 30nM KCl or 100nM capsaicin. We only analyzed the L1-L3 DRG DiI+ neurons which were supposed to represent the neurons innervating the laminectomy area at the lower back region 7 . And only the calcium transients of the potassium responsive neurons were analyzed.
  • 20. On POD1, laminectomy potentiates K+ evoked Ca2+ transients in DRG neurons and RTX attenuated the Ca2+ transients enhancement induced by laminectomy. The magnitudes of K+ evoked Ca2+ transient were analyzed using the change of peak ratio to baseline. And the decay was measured as time from onset to return to 20% of peak (T80). Results of this analysis for DiI+ neurons revealed significant effects associated with laminectomy (P<0.001). When using K+ to depolarize the DRG neurons, both the magnitude and the decay of Ca2+ transients in DRG neurons were enhanced by laminectomy. Laminectomy induced higher amplitude (laminectomy 1.70±0.07vs. sham 1.26±0.09, ***p<0.001, Fig.1) and longer decay (laminectomy 269.21±34.98 vs. sham 137.6±15.55, ***p<0.001, Fig.2) when compared with sham group. Laminectomy significantly enhanced the Ca2+ transients evoked by 30nM KCl depolarization and attenuated the recovery from depolarization evoked by 30nM KCl.To determine whether RTX can have effect on DRG neurons’ K+ evoked Ca2+ transients, we administered 200ng RTX intrathecally at L4 and L5 level 7 days before laminectomy.Post-hoc (Tukey)analysis indicated that intrathecal 200ng RTX pretreatment reduced the K+ evoked calcium flux in laminectomy group. (laminectomy 1.70±0.07, vs. RTX and laminectomy 1.14±0.07, *** p<0.001, n= 46 vs 31 cells, 6 rats, Fig. 1), while the decay of the K+ evoked Ca2+ transient was shortened by RTX pretreatment (laminectomy 269.2±34.98, vs. RTX and laminectomy 82.43±11.12, *** p<0.001, n= 25 vs 21cells, 6 rats, ***P<0.001, Fig. 2). Thus, potassium-depolarization mediated calcium mobilization in laminectomy group was drastically reduced in the presence of RTX. On POD1, RTX also can reduce the amplitude of calcium flux in sham rat DRG neurons. (sham 1.26±0.09, vs. RTX and sham 0.88±0.07, **p<0.01, n= 45 vs
  • 21. 44cells, 6 rats, Fig.1). and shortened the duration. (sham 137.7±15.55, vs. RTX and sham 74.46±8.29, *p<0.05, n= 45,37 cells, 6 rats, Fig.2). Therefore, on POD1, in both laminectomy rats and sham rats, RTX reduced the calcium response to KCl , either by reducing the amplitude of calcium peak or by shortening the decay of the transient. On POD1, capsaicin evoked Ca2+ transients in DRG neurons was also potentiated by laminectomy, which was attenuated by intrathecal RTX. Laminectomy induced higher amplitude when compared with sham group (laminectomy 2.60±0.14 vs. sham 2.03±0.14, *p<0.05, Fig. 3 Post hoc analysis (Tukey test)) and slow the recovery of transients in small diameter neurons (laminectomy 986.10±116.10, vs. sham 511.31±78.65, ***P<0.001, Fig. 4). And there is also a significant increase in the percentage of capsaicin responsive neurons in the DRG populations induced by laminectomy (laminectomy 75.20±3.79% vs. sham 63.67±1.93%, *P<0.05, Fig. 5). As observed RTX effect on potassium induced calcium transients, there was also a statistically significant effect of pre-surgery RTX in both the magnitude (laminectomy 2.60±0.14, vs. RTX and laminectomy 1.52±0.16, *** p<0.001, n= 40 vs 34 cells, 6 rats, Fig. 3)and decay (laminectomy 986.10±116.10, vs. RTX and laminectomy 378.70±69.18, *** p<0.001, n= 32 vs 27 cells, 6 rats, Fig. 4) of the capsaicin evoked Ca2+ transient in small DiI+ DRG neurons. In addition, RTX decreased the percentage of capsaicin responsive DRG (laminectomy 75.20±3.79% vs. RTX 51.00±3.67%, ***p<0.001, Fig. 5). It also decreased the percentage of capsaicin responsive DRG in sham rats (sham 63.67±1.93% vs. RTX and sham 33.10±1.78%, ****P<0.0001).
  • 22. On POD1, in both laminectomy rats and sham rats, RTX reduced the calcium response to capsaicin, either by completely eliminating the transient or by reducing the amplitude of calcium peak or by shortening the decay of the transient. On POD7, laminectomy did not produce significant changes in K+ evoked Ca2+ transients in DRG neurons either in amplitude (laminectomy 1.04±0.11vs. sham 1.18±0.08, p>0.05,n= 23 vs 40 cells, 4 rats Fig. 6) or decay(laminectomy 108.70±26.31 vs. sham 116.2±12.79, p>0.05, Fig. 7). On POD7, laminectomy extended the decay of capsaicin evoked Ca2+ transients. And RTX attenuated the laminectomy enhanced capsaicin response in DRG neurons On POD7, even the magnitude of capsaicin evoked Ca2+ transients did not differ between laminectomy group and sham group(laminectomy 2.19±0.18 vs. sham 1.77±0.15,p>0.05, n=20 vs 38 cells, 4 rats each group Fig. 8) although RTX decrease the amplitude both in laminectomy rats (laminectomy 2.19±0.18, vs. RTX and laminectomy 1.21±0.12, n=20 vs 37 cells , 4 rats each group,*** p<0.001, Fig. 8) and sham rats(sham 1.77±0.15% vs RTX and sham 0.85±0.09%, n=38 vs 30 cells, 4 rats each group, ***P<0.001, Fig. 8). However there was still increased decay of capsaicin evoked Ca2+ transients in laminectomy group (laminectomy 1044±149 vs sham 414±44.76 seconds, n= 25vs 33 cells, 4 rats each group, ***P<0.001, Fig. 9), and RTX shortened the decay in both laminectomy rat (laminectomy 1044.0±149.3 vs RTX and laminectomy 188.6±33.68
  • 23. seconds, n=25 vs 26 cells, 4 rats each group, ****P<0.0001, Fig. 9) and sham rats(sham 414±44.76% vs RTX and sham 161.2±11.89%, n=33 vs 35 cells, 4 rats each group, *P<0.05, Fig. 9). On POD7, there was no significant difference in terms of the percentage of capsaicin responsive neurons between the sham group and laminectomy group (laminectomy 71.75±6.96% vs. sham 59.51±2.82%, 4 rats each group, p>0.05, Fig. 10), even when RTX still lowered the percentage of capsaicin responsive DRG neurons both in RTX pretreated laminectomy group (laminectomy 71.75±6.69% vs RTX laminectomy 45.0±2.04%, 4 rats each group,**p<0.01, Fig. 10) and sham rats(sham 59.51±2.82% vs RTX and sham 33.60±1.50%, n=10 vs 5 rats each group, *P<0.001, Fig. 10). Thus, on POD7, RTX still attenuated the laminectomy enhanced DRG neurons’ responses to capsaicin. Effects of RTX on wheel running and home cage activity We observed the whole week after laminectomy using RTX. And found significant changes after RTX administration. The attenuated behavior was recovered faster compared with laminectomy alone. Including running distance and locomotion.
  • 24. rtx la m i D is ta n c e B a s e lin e 0 1 2 3 4 5 6 7 0 5 0 1 0 0 1 5 0 2 0 0 R T x + L a m in e cto m y lia n g N o D ru g (N o A lz e t) L a m i lia n g S h a m lia n g rtx la m i M a x S p e e d B a s e lin e 0 1 2 3 4 5 6 7 0 5 0 1 0 0 1 5 0 R T x + L a m in e cto m y L a m i S h am wo     On  pod6  7  there  is  a  sig  diff  between  RTX  and  no  drug  laminectomy  in  running  distance  (two  way  anova,   for  multiple  t-­‐  test,  day  3,4,5,6,7  is  different.  P=0.016,  0.0013,0.0013,0.0002,0.0004).  On  pod0,  there  is  a   diff  between  RTX  and  no  drug  in  maximum  speed(two  way  anova,multiple  t  test,  day  0,1,2,   p=0.0003,0.01,0.02).  there  is  no  diff  between  the  2  group  in  acceleration.  
  • 25. R T X F in e m o to r m o v e m e n t B a s e lin e 0 1 2 3 4 5 6 7 0 5 0 1 0 0 1 5 0 S h am L a m in e c to m y L a m in e cto m y -R T x R T X R e a rin g B a s e lin e 0 1 2 3 4 5 6 7 0 5 0 1 0 0 1 5 0 S h a m lia n g L a m in e c to m y L a m in e cto m y R T x
  • 26. there is a sig diff between RTX and no drug laminectomy in fine motor movement (two way anova, for multiple t- test, day 1,2 is different. P=0.014, 0.026). there is a diff between RTX and no drug in rearing(two way anova,multiple t test, day 1, p=0.008). there is no diff between the 2 group in acceleration. Effects of RTX on the thermal hyperalgesia The effects of the intrathecal administration of RTX (200ng) on thermal hyperalgesia in naïve rats at 7 days and 10 days after injection of RTX were investigated. Our results show that the intrathecal administration of 200ng RTX inhibited the thermal response of hindpaw (Fig. 11), which confirmed the valid intrathecal injection of RTX. Discussion The pain after operation represents a significant clinical challenge for treatment. The search for new treatment options of postoperative pain has been directed toward specific mediators and modulators of pain and inflammation. In this study, we showed that laminectomy sensitized the DRG sensory neurons indicated by elevated amplitude of calcium response curve and prolonged duration both in response to high K and capsaicin and a higher percentage of capsaicin responsive ,TRPV1 positive neurons. The present study also showed that it RTX attenuated DRG responsiveness to potassium and capsaicin evidenced by recovered amplitude, reversion of prolonged duration to high K and capsaicin and reduced percentage of TRPv1 positive DRG neurons.
  • 27. Furthermore, on pod7 laminectomy still has prolonged the duration of intracellular neuronal calcium response to capsaicin, which is also reversed by pretreatment of i.t. RTX indicated by a normalized duration of calcium response. This showed an extended effect of RTX. Laminectomy produces a decrease in rat wheel running and spontaneous home cage activities. I .T. RTX pretreatment reversed the decrease of wheel running distance and recovered the rearing and fine movement. Because there were no sided effects observed in the experiment, thus the safety profile of RTX was established. We propose that RTX intrathecal application could be a useful strategy in alleviating postoperative pain. We developed an rat model of postoperative pain and examined whether TRPV1 play a role in mediating postoperative pain injury. In sensory dorsal root ganglia, the trpv1 are expressed on small and medium –sized neurons and are mainly involved with mediation of nociceptive information. TRPV1 is expressed by the type II A-delta and C-fiber neurons, functioning as a molecular integrator of nociception. TRPV1-positive neurons are essential for the development of mechanical allodynia. Trvp1 is sensitized by phosphorylation of trpv1 via cAMP8 . and we tested he effect of specific TRPV1 agonists could overexcited DRG neurons and provide a potential way of relieving pain. Prior studies have shown that pain could induce expression of TRPV1 and neuron phenotype change. Nerve injury and inflammation could enhance the expression of
  • 28. trpv1 including the transcription, translation, post translational modification and trafficking of trpv1(Hucho T 2007 neuron, zhang x, EMBO J 2005, Ji RR, Neuron 2002). The net effect is central and peripheral sensitization. This could switch the phenotype of DRG nruosns and spinal cord neurons from naïve state to an altered state. Thus redcuce the pain threshold(Patapoutian A, 2009).Trpv1 could enhanced form gene level, translational level (mRNA), and post-translational modifications such as phosphorylation, n-glycosylation and covalent modification9 . Especially phosphorylation by PKCᵋ (Cesare 1999, Mandadi S 2006 pain) 10 and PKA(Bhave 2002, Lopshire, 1998). Also there is evidence showing the phosphorylation of Trpv1 is enhanced. .Inflammaorty mediators sensitized trpv1 through phosphorylation of trpv1 by protein kinase PKCᵋ (Mandadi S 2006 Pain zhang H, 2007 J neuroscience). In postoperative rat, trpv1 could be elevated because of gene level, rna level and posttranstlational level change. Recent study demonstrated that TRpv1 antagonist intratheally and local application both can allevievate thermal hyperalgsiea and mechanical allodynia(Uchytilova mole pain ,2014 pure behavior). Also in a model of thoracotomy postoperative pain, RTX significantly reduced mechanical allodynia following surgery.(Shin Jw , 2010). TRPV1-expressing C-fibers are sensitized to heat and acid after incision (Kang S 2010 Pain), All this indicated the involvement of trpv 1 in postoperative pain. As we have demonstrated here, there were marked differences in small DRG neurons between laminectomy group and sham group with respect to the magnitude and decay of capsaicin-evoked Calcium transients. In order to identify neurons innervating the site of surgery, DiI was injected in lumbar region prior to the
  • 29. surgery. One day after surgery, there was a significant increase in the magnitude and decrease in the decay of capsaicin evoked calcium transients in small DRG neurons. Similar decreases in decay were also observed in the transients evoked by capsaicin even 7 days after laminectomy. The reason of the enhanced intracellular calcium transients during inflammation could be the inflammatory mediators such as nerve growth factor because they might affect the calcium efflux, uptake and binding of calcium and thus contribute to the enhanced calcium transients during inflammation (Lu, 2008 ). The trpv1 sensitaztion in laminectomy group could be attributed to inflammatory mediators such as Nerve growth factor (NGF) because it induces sensitization of the TRPV1(Zhu W, 2007 mol cell neurosci). Other Inflammatory mediators (BK, substance P) also modulate TRPV1 activity (Sugiura T. Bradykinin lowers the threshold temperature for heat activation of vanilloid receptor 1. J Neurophysiol 2002; 88:544–8. Neurokinin-1 receptor enhances TRPV1 activity in primary sensory neurons via PKCepsilon: a novel pathway for heat hyperalgesia. J Neurosci. 2007 Zhang H). Studies have shown that trpv1 sensitization(by heat) produced a larger membrane deploarizations in rat DRGs. This could lower the threshold and increase excitability of DRG neurons. (Neelands, 2008 inflam research, zhang xf, 2011). We demonstrated that surgery induced an enhanced calcium response to both potassium and capsaicin. Because Intracellular calcium plays a critical role in mediation of many cellular processes such as transcription, ion channel activity, it regulates the neuronal excitability and transmitter release (Berridge, 2005)and hormone release, and Ca2 + -dependent gene transcription. , so the enhanced calcium response seen in small DRG after surgery might contribute to the postoperative pain by changing
  • 30. afferent excitability and facilitating transmitter transcription and release from the nociceptive afferents. Indeed, pervious study showed that activation of trpv1 in afferent terminals could increase the rate of glutamate release by allowing calcium into neurons (Shoudai 2010). Such results suggest that activated, presynaptic TRPV1+ receptors trigger continuous resting release of glutamate vesicles at physiological temperatures only in capsaicin-responsive terminals. Thus the sensitization of DRGs found in postoperative pain model indicated the higher excitability of DRG neurons and increased neurotransmitter release in afferent terminals. In terms of percentages of trpv1 response positive cells, unregulated trpv1 responsiveness in primary-afferent fibers occurs in inflammatory disease and irritable bowel syndrome (yiangou 2001, Chan 2003). Perivous study has shown that nerve injure leads to immnunoreactivity changes from trpv1 negative to positive in large diameter neurons after nerve injury(Hudson 2001). A similar gain of capsaicin sensitivity was seen in calcium imaging recently after nerve injury (Kim 2014 neuron). These phenotypic switch was mostly seen in nerve injury and chronic pain(Costigan M 2009 Annu Rev Neurosci). Interestingly, cancer pain also leads to increase of trpv1 expression in both myelinated DRG neurons and perptidergic unmyelinated DRG neurons (Niiyama Y, 2007 neuroscience). In our experiment, the intracellular calcium response to capsaicin is also enhanced in terms of percentages of positive cells. The percentage of TRPV1 responsive DRG rose in pain group was higher compared with control group. The percentage change might reflect a phenotype change in DRG population. This findings extended previous research results by showing that
  • 31. unregulated trpv1 responsiveness in primary-afferent fibers occurs in postoperative pain, Together, these data are consistent with the hypothesis that the TRPV1 contribute to postoperative pain DRG excitability. Indeed, the responses are both elevated in pod1 and pod7 when stimulated with capsaicin. To further understand the role to trpv1- positive neurons in development of postoperative pain, we studied the Pod7 response in DRGS. On PoD7, We found the the amplitude is reduced and d the percentage is recovered. The amplitudes of capsaicin evoked calcium transients and percentages of capsaicin responsive neurons are not higher in surgery group when compared with control. This showed that the surgery induced a recoverable enhanced response in DRGS indicating the disappearance of the inflammaotory part of postoperative pain. However on pod7 the duration of capsaicin induced calcium response remains longer in surgery group. This could due to the long- lasting remaining sensitization of DRGS after surgery. This extended effect of surgery on calcium responses was reflected by our wheel running results, which showed that on Pod7 there is still a difference between RTX and no treatment group in running distance(On pod6 and pod7 there is a sig diff between RTX and no drug laminectomy in running distance (two way anova, for multiple t- test, day 3,4,5,6,7 is different. P=0.016, 0.0013,0.0013,0.0002,0.0004). In our study, One day after the laminectomy, there was also a significant increase in the magnitude and decrease in the decay of potassium evoked calcium transients in DiI labeled neurons. We found the response duration, amplitude of DRG response to k30 to be elevated on POD1. And this enhanced response disappeared on POD7. Evidence
  • 32. showed that the nerve injury in DRG leads to decrease in resting calcium concentration (Fuchs ,2005) and a decrease in both the magnitude and decay of evoked calcium transients (Fuchs 2007)and nerve injury also leads to a decreased voltage gated calcium currents in injured DRG (MaCallum 2003, Baccei and Kocsis,2000). Also there was reports that inflammation is associated with increases in magnitude and decrease in decay of the high potassium evoked transient in small putative nociceptive afferents(Lu 2008,neuroscience). In the present postoperative pain model, high potassium induced weak excitation of naïve DRG , but strongly activate the DRG in laminectomy group as indicated by increase in calcium transients’ amplitudes and durations. This augmented responsiveness indicated neuronal sensitization. This stronger activation is possibly due to trpv1 sensitization in laminectomy group because inflammatory mediators induces an acute sensitization of nociceptive DRG neurons partially through sensitization of the TRPV1(Zhu W, 2007 mol cell neurosci). The high responsiveness to KCl and capsaicin shown in our postoperative pain model support the idea of sensitization of DRG and phenotypic changes in DRGs after surgery. This enhanced responsiveness to potassium disappeared on POD7. Using this pain model, we also showed that the significantly augmented DRG neurons’ response to both potassium and capsaicin challenges are blunted by the application of TPV1 agonist RTX. Also the percentage changes happened following the surgery are also prevented by pretreatment of RTX. RTX mediated antinociceptive effects both in home cage locomotion and wheel running. Trpv1 agonists-induced analgesia is proposed be due to receptor desensitization because of a sustained activation of
  • 33. receptors()Jeffry Jap los one 2009, Raisinghani M 2005 j physiol). I. t. RTX couls cause aa sustained increased freuqencey of spontaneous EPSCs and lead to a desensitization response and depolarization block, synaptic failures(Jeffry Ja plosone 2009 sikand p, 2007 j physiol).. RTX has been shown to cause both spinal cord and DRG changes if applied intrathecal. RTX could induce a degenaeraion of trpv1 positive neurons by causing a neurotoxic influx of calcium ion through the channels resulting a calcium induced neuronal death(karai 2004). Our data extended the previous findings by showing that the percentage of DRG neurons responveis to capsicien is significantly reduced aftet RTX treatment. Also it has been shown that the ablation of central terminals of trv1 expressing neurons occured. after i.t. RTX (Bishinoi M 2011 j pain). Even when used perineurally and remote from the neuronal perikary in DRG , RTX still could ablate DRG TRPV1 positive neurons (Neubert 2008). RTX is neuroexcitotoxic and ablate neurons. RTX is a promising drug for treatment because of its desensitizing effect and ablate effect. Single dose might desensitize ,repeated dose or high dose could cause neurotoxity and permanently degenerate nerve Trpvv1 + nerve endings (Ladarola 2011). RTX induced a prolonged increase in intracellular calcium in Trvp1 positive neurons in human dorsal root ganglion cultures, , while leaving other adjacent neurons unaffected(Karai, 2004). It has been proved that intrathecal RTX could reduce TRpv1, CGRP and trpa1 immunoreactivities in the lumbar dorsal root ganglia and (karai 2004, Neubert 2008 Klafke 2012 neuroscience), indicating the deltetion ot DRG TRPV1- expresssing neurons(Neubert). Intrathecal rtx also produce ablation of central nerve
  • 34. terminals of DRG neurons indicated by decreased immune reaction of TRV1 in spinal cord dorsal horn (Bishnoi 2011). This cellular changes were reflectd in behavioral tests such as prolonged hind paw thermal stimulation latency and carrageenan-induced inflammatory therma lanalgesia(Bishnoi 2011). Our thermal test on hind paw confirmed the prolonged radiant noxiouls thermal stimulus latency and suggested the specific deletion of trpv1 positive neurons. In the present study, intrathecal RTX reduced the calcium response. We found that RTX producing a significantly reduced responsiveness to potassium and capsacin in DRGs.Also In calcium imaging, the percentages of trpv1 responsive DRG neurons decrease after RTX application both in postoperative rats and sham rats further proved that the intrathecal RTX targeted trpv1 positive neurons in DRG and made them inert to high potassium and capsaicin. We propose that i.t. RTX also affect DRG trpv1 channels because RTX diffused to DRG and act on primary afferent neurons. This may partly explained our finding that after rtx, the DRG response to capsaicin are reduced. This further extended the findings that RTX decreasing the TRPV1 immunoreactivities in DRG after both perineural and intrathecal RTX application(neubert Jk, 2008, Klafke 2012 neuroscience) RTX has been used for debilitating arthritic and cancer pain . In rats of postoperative pain, RTX can significantly reduce the pain(my paper). Also the RTX has been shown to be effective in treatment of neuropathic pain(gariel Tender, spinaljournal 2008) because RTX jinjection acuse an excitotoxic effect when injected into sensory ganglia. RTX is a
  • 35. transient receptor potential vanilloid 1 channel agonist. It also contributes to inflammatory and heat allodynia relieve. Our intrathecal injection of RTX also cause an inert DRG neuron reaction to K30 and capsaicin, both on pod1 and pod7. Extensive research dta have shown that trpV1 positive neurons mediate thermal hyperalgesia in pain models.(11-16 of Tender 2008 spine ournal). Moreover , the same cells apppeart to be responsible ofre the mediation of neurogenic inflamesion(13.17-19), blockage of trpv1 apears eefective in reversing established hyperalgesia associate with inflammation(18). Our data corroborated that RTX could abolish the heat induce pain in rats, evidence by the extenede time response time to the heat application on hindpaw in rats treated with i.t. RTX. The paw became less responsive to heat. These results are in concordance with previous studies showing theat trpv1-posticitve neurons mediated termal hyperalgesia. Also it is shown that inhibition of trpv1 centrally could inhibit the mechanical hyperalgesia even it is not a mechanotransducer(Kim 2014 Neuron). So far there have been no data to demonstrate the involvement of trpv1-positive neurons in mechanical sensitivity or allodynia. However, some earlier reports showed that RTX injected in DRGs could elevated the average withdrawal threshold of the paws and the magnitude of touch –evoked allodynia was improved, which means the RTX improved tactile allodynia via selectively eliminating the trv1-positive cells in the DRGs. This could be explained by the cross-talk between A-beta, A-delta and C-fiber neurons in DRGs (23 of
  • 36. tender 2008) . After operation, large amounts of glutamate and aspartate are released into extracellular space and the inhibitory neurotransmitter GABA is reduced in spinal dorsal horn (22) and also in the DRGs, resulting the hypersensitivity of A fibers and C fibers. When use a touch test of allodynia, the A-beta fibers carried the signals to the DRG and excited not only the A-beta cell bodies but also A-delta and C neurons because of the cross-excitation. The excitation of a A-delta and C neurons subsequently contribute to the touch induced pain and personal’s experience and emotional status. Thus, deletion of trpv1 neurons (include A-delta and C fibers) could reduce the excitation of A-delta and C- fibers in DRGs and thus reduce the whole animal’s response to touch. Also even we did not test the mechanical threshold of back region. There was evidence showing that the surgery affected the cutaneous sensitivity in the back region after thoracotomy and RTX could relieve the allodynia developed after(Shin 2010) surgery11 . It can relieve pain like shown in behavioral tests wheel running and spontaneous activities. It already has been shown when used in large doses intrathecally(3.8ug/kg), it is able to alleviate capsaicin induced nocifensive behavior(bishnoi 2011 j pain) and both capsaicin and carrageenan induced inflame thermal hypersensitivity  (bishnoi 2011 j pain).    Also it is shown that inhibition of trpv1 centrally could inhibit the mechanical hyperalgesia even it is not a mechanotransducer(Kim 2014 Neuron). We observed the spontaneous activity after surgery and the wheel running behavior after laminectomy instead, it is shown that the wheel running and home cage activity are changed by laminectomy and recovered partially due to RTX. We say the RTX
  • 37. improved the running distance and the spontaneous activity in postoperative rats. Such as rearing and fine movements in homecage, which proved that RTX mediated antinociceptive effects and even after intrathecal RTX, animal maintain the motor control. There was report that RTX caused a long lasting analgesia in cancer pain model even up to 14 week after application and the hind paw heat response was significantly reduced (Brown 2005). Our heat test further confirmed that intrathecal RTX induced a higher heat response threshold. Also it has been shown that lumbar epidural application of RTX resulted a focused distribution to lumbar neurons and lead to a profound and segmental analgesia12 . Recently it was shown that intrathecal RTX could lead to dose-dependent deletion of TRPV1 CGRP and phosphor ERK immunoreactivities in superficial spinal cord such as laminae I and II. Other studies showed the physiological sensation could recover 2 weeks after RTx treatment2 . In summary, the high responsiveness to capsaicin and potassium in DRG reflected the sensitization of neurons and this indicates high excitability and more neurotransmitter release in peripheral nociceptive neurons. RTX is a promising pain killer because both the calicium imaging and behavioral test support its use in postoperative pain. 1.   Mishra  SK,  Hoon  MA:  Ablation  of  TrpV1  neurons  reveals  their  selective  role  in  thermal  pain   sensation.  Mol  Cell  Neurosci  2010;  43:  157-­‐63   2.   Neubert  JK,  Mannes  AJ,  Karai  LJ,  Jenkins  AC,  Zawatski  L,  Abu-­‐Asab  M,  Iadarola  MJ:  Perineural   resiniferatoxin  selectively  inhibits  inflammatory  hyperalgesia.  Mol  Pain  2008;  4:  3  
  • 38. 3.   Karai  L,  Brown  DC,  Mannes  AJ,  Connelly  ST,  Brown  J,  Gandal  M,  Wellisch  OM,  Neubert  JK,  Olah  Z,   Iadarola  MJ:  Deletion  of  vanilloid  receptor  1-­‐expressing  primary  afferent  neurons  for  pain  control.  J  Clin   Invest  2004;  113:  1344-­‐52   4.   Zimmermann  M:  Ethical  guidelines  for  investigations  of  experimental  pain  in  conscious  animals.   Pain  1983;  16:  109-­‐10   5.   Lu  SG,  Zhang  X,  Gold  MS:  Intracellular  calcium  regulation  among  subpopulations  of  rat  dorsal   root  ganglion  neurons.  J  Physiol  2006;  577:  169-­‐90   6.   Waxman  SG,  Dib-­‐Hajj  S,  Cummins  TR,  Black  JA:  Sodium  channels  and  pain.  Proc  Natl  Acad  Sci  U  S   A  1999;  96:  7635-­‐9   7.   Takahashi  Y,  Chiba  T,  Kurokawa  M,  Aoki  Y:  Dermatomes  and  the  central  organization  of   dermatomes  and  body  surface  regions  in  the  spinal  cord  dorsal  horn  in  rats.  J  Comp  Neurol  2003;  462:   29-­‐41   8.   Bhave  G,  Zhu  W,  Wang  H,  Brasier  DJ,  Oxford  GS,  Gereau  RWt:  cAMP-­‐dependent  protein  kinase   regulates  desensitization  of  the  capsaicin  receptor  (VR1)  by  direct  phosphorylation.  Neuron  2002;  35:   721-­‐31   9.   Voolstra  O,  Huber  A:  Post-­‐Translational  Modifications  of  TRP  Channels.  Cells  2014;  3:  258-­‐87   10.   Cesare  P,  Dekker  LV,  Sardini  A,  Parker  PJ,  McNaughton  PA:  Specific  involvement  of  PKC-­‐epsilon   in  sensitization  of  the  neuronal  response  to  painful  heat.  Neuron  1999;  23:  617-­‐24   11.   Shin  JW,  Pancaro  C,  Wang  CF,  Gerner  P:  The  effects  of  resiniferatoxin  in  an  experimental  rat   thoracotomy  model.  Anesth  Analg  2010;  110:  228-­‐32   12.   Szabo  T,  Olah  Z,  Iadarola  MJ,  Blumberg  PM:  Epidural  resiniferatoxin  induced  prolonged  regional   analgesia  to  pain.  Brain  Res  1999;  840:  92-­‐8