Você está na página 1de 7

Cellular, molecular, and developmental neuroscience 1

Evaluation of apoptotic pathways in dorsal root ganglion


neurons following peripheral nerve injury
Rebecca Wiberga,b, Liudmila N. Novikovaa and Paul J. Kinghama

Peripheral nerve injuries induce significant sensory associated with isolectin B4-positive neurons in the DRG
neuronal cell death in the dorsal root ganglia (DRG); and this may provide an explanation for the increased
however, the role of specific apoptotic pathways is still susceptibility of these neurons to die following nerve injury,
unclear. In this study, we performed peripheral nerve likely in part because of an activation of the ER-stress
transection on adult rats, after which the corresponding response. NeuroReport 00:000–000 Copyright © 2018
DRGs were harvested at 7, 14, and 28 days after injury for Wolters Kluwer Health, Inc. All rights reserved.
subsequent molecular analyses with quantitative reverse NeuroReport 2018, 00:000–000
transcription-PCR, western blotting, and
immunohistochemistry. Nerve injury led to increased levels Keywords: apoptosis, endoplasmic reticulum, peripheral nerve injury,
sensory neurons
of caspase-3 mRNA and active caspase-3 protein in the
a
DRG. Increased expression of caspase-8, caspase-12, Department of Integrative Medical Biology, Section of Anatomy and bDepartment
of Surgical & Perioperative Sciences, Section of Hand and Plastic Surgery, Umeå
caspase-7, and calpain suggested that both the extrinsic University, Umeå, Sweden
and the endoplasmic reticulum (ER) stress-mediated
Correspondence to Rebecca Wiberg, PhD, Department of Integrative Medical
apoptotic pathways were activated. Phosphorylation of Biology, Section of Anatomy, Umeå University, SE-901 87 Umeå, Sweden
protein kinase R-like ER kinase further implied the Tel: + 46 90 786 9754; fax: + 46 90 786 5480; e-mail: rebecka.wiberg@umu.se

involvement of ER-stress in the DRG. Phosphorylated Received 23 February 2018 accepted 20 March 2018
protein kinase R-like ER kinase was most commonly

Introduction activation of cysteine-dependent aspartate-directed pro-


Peripheral nerve injury induces significant sensory neu- teases, the caspases. Three main different kinds of
ronal cell death in the dorsal root ganglia (DRG). In apoptosis exist [6,7]: (i) the intrinsic apoptotic pathway,
contrast to spinal motoneurons, both neonatal and adult which is regulated by the balance between proapoptotic
primary sensory neurons in the DRG undergo cell death Bax and antiapoptotic Bcl-2, acting at the mitochondrial
even after distal peripheral nerve injury [1], which is also level determining the membrane permeabilization, (ii)
the case for cranial motoneurons [2]. Moreover, cuta- the extrinsic apoptotic pathway, which is characterized by
neous afferent neurons are more sensitive to injury- the presence of death receptors (DRs), and (iii) apoptosis
induced degeneration in comparison with muscular mediated by endoplasmic reticulum (ER) stress, with
afferents [1,3,4]. Sensory neuronal cell death starts each pathway culminating in cleavage of the effector
shortly after nerve axotomy and experimentally it has caspase-3.
been shown that a significant cell loss is already evident In this study, we used an animal model of peripheral
in the DRG 1 week after injury if repair is not carried out nerve injury to gain a better understanding of the apop-
[5]. Loss of sensory neurons increases with time, reaching totic mechanisms underlying sensory neuronal cell death
a value of 35–40% of the total neuronal cell population in in the DRG. We have analyzed the expression of
the DRG at 2 months after injury [5]. Despite advanced caspase-3, caspase-8, and caspase-12 as respective med-
microsurgical innovation, immediate epineural nerve iators of the intrinsic, extrinsic, and ER-stress apoptotic
repair only mediates partial neuroprotection, reducing pathways.
the amount of neuronal cell loss from 21 to 5% 2 weeks
postoperatively [5], creating a need for an additional
pharmacological intervention. Material and methods
Experimental animals and ethics statement
Current data on the underlying mechanism behind Adult (10–12 weeks old) female Sprague-Dawley rats
injury-induced cell death are conflicting, with uncer- with a mean weight of 250 g were used in this study. The
tainties with respect to the specific apoptotic pathways animal husbandry was in accordance to the standards and
involved. Apoptosis consists of several biochemical regulations provided by the National Institutes of Health
events leading to characteristic morphological changes, Guide for Care and Use of Laboratory Animals (NIH
including membrane blebbing, cell shrinkage, nuclear publications no. 86–23, revised 1985) and the European
fragmentation, chromatin condensation, and DNA frag- Communities Council Directive (86/609/EEC). All pro-
mentation, ultimately resulting in cell death because of cedures were approved by the Northern Swedish
0959-4965 Copyright © 2018 Wolters Kluwer Health, Inc. All rights reserved. DOI: 10.1097/WNR.0000000000001031

Copyright r 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
2 NeuroReport 2018, Vol 00 No 00

Regional Committee for Ethics in Animal Experiments cDNA synthesis kit). Quantitative RT-PCR was subse-
(DNR #A186-12). Surgery was performed aseptically quently performed using SsoFast EvaGreen Supermix
using general anesthesia consisting of a mixture of keta- (BioRad, Solna, Sweden) according to the manufacturer’s
mine (Ketalar 50 mg/ml; Pfizer, Strängnäs, Sweden) and recommendations. Caspase-3: forward: 5′-GGACCTGT
xylazine (Rompun 20 mg/ml; Bayer Healthcare, Berlin, GGACCTGAAAAA-3′ and reverse: 5′-AGTTCGGCTT
Germany) by an intraperitoneal injection. The well-being TCCAGTCAG-3′ primers with an annealing tempera-
of the rats was observed throughout the experimental ture (Ta) of 60.5°C, caspase-8: forward: 5′-CTGGGAAG
period. GATCGACGATTA-3′ and reverse: 5′-CATGTCCTGC
AGTTTTGATGG-3′ primers with Ta of 61°C, caspase-
Surgical procedures and experimental groups 12: forward: 5′-GGCAGACATACTGGTACTATTTG
The animals were divided into four groups; sciatic nerve G-3′ and reverse: 5′-GCTCAACACACATTCCTCATC
transection without repair with 7 (n = 5), 14 (n = 5), and 28 TGT-3′ primers with Ta of 61°C, caspase-7: forward:
(n = 5) days of survival, and a control group consisting of 5′-TACAAGATCCCGGTGGAAGCT-3′ and reverse:
unoperated animals (n = 9). A separate series of animals 5′-CTGGGTTCCTCCACGAATAAT-3′ primers with
was used for immunohistochemisty (n = 3) and for wes- Ta of 62°C, calpain: forward: 5′-TGCTCTGCCGAAGT
tern blotting (n = 4–6/group). The left sciatic nerve was GTCACC-3′ and reverse: 5′-GCACTGGATGGCCT
exposed by bluntly dividing the gluteal muscles of the GGAGTT-3′ primers with Ta of 66°C, and the reference
thigh. Under an operating microscope (Zeiss; Carl Zeiss, gene 18s: forward: 5′-TCAACTTTCGATGGTAGTC
Oberkochen, Germany), the nerve was transected at a GC-3′ and reverse: 5′-CCTCCAATGGATCCTCGTT
standardized distance from the spinal cord ∼ 5 mm AA-3′ primers with Ta of 62°C were purchased from
proximal of the sciatic nerve’s first nerve branch. The Sigma (Poole, UK).
nerve stumps were then capped to prevent distal rein-
nervation. Caps were made out of polyethylene tubes Western blotting
and each nerve stump was introduced and anchored to L4, L5, and L6 DRGs were dissected from nerve-
the cap using the 10-0 Ethilon suture. After surgery, the transected animals after 7, 14, and 28 days without
wound was closed in layers: muscles with a 3-0 Silk repair and also uninjured control animals. DRGs were
suture and the skin with a 3-0 Silk suture. The operated pooled in each group and prepared by homogenization in
animals were allowed to survive for 7, 14, and 28 days protein lysis buffer. Whole-tissue lysates were analyzed
before harvesting. for protein content using a commercially available protein
assay kit (BioRad). The antibodies used were rabbit anti-
Tissue processing caspase-3 antibody (1 : 1000, Catalog Number 9662;
At the end of the survival period, the rats were injected Cell Signaling Technology, BioNordika, Stockholm,
terminally with an intraperitoneal overdose of sodium Sweden), mouse anti-caspase-7 antibody (1 : 500, Catalog
pentobarbital (240 mg/kg; Apoteksbolaget, Stockholm, Number ab2301; Abcam, Cambridge, UK), rabbit anti-
Sweden). For reverse transcription-PCR (RT-PCR) and caspase-12 antibody (1 : 500, Catalog Number ab62484;
western blotting, the L4, L5, and L6 DRGs from the Abcam), rabbit anti-caspase-8 antibody (1 : 1000, Catalog
operated side were harvested and fast frozen in liquid Number NB100-56116; , Novus Biologicals, Abingdon,
nitrogen. For immunohistochemistry, the animals were UK), or rabbit anti-phospho-protein kinase R-like endo-
transcardially perfused with Tyrode’s solution, followed plasmic reticulum kinase (anti-phospho-PERK) (1 : 1000,
by 4% (w/v) paraformaldehyde in 0.1 M PBS (pH 7.4). Catalog Number 3179; Cell Signaling Technology). Anti-
The L4, L5, and L6 DRGs were harvested, postfixed for β-tubulin antibody (1 : 1000, Catalog Number ab6046;
2–3 h, cryoprotected in 20 and 30% sucrose for 2–3 days, Abcam) was used as the loading control antibody.
and frozen in liquid isopentane. Serial transverse 16-μm
thick sections were cut on a cryomicrotome (Leica Immunohistochemistry
Instruments, Kista, Sweden), thaw-mounted in pairs onto A 16-μm thick L4, L5, and L6 DRG sections were
SuperFrost Plus slides, dried overnight at room tem- blocked with normal serum, after which the following
perature, and stored at − 80°C before processing. primary antibodies were used: anti-active caspase-3
antibody (1 : 10, Catalog Number ab2302; Abcam), rab-
Quantitative reverse transcription-PCR bit anti-phospho-PERK (1 : 200, Catalog Number sc-32577;
Total RNA was isolated from DRGs using a miRNeasy Santa Cruz Biotechnology, Heidelberg, Germany), and
mini kit (Qiagen, Sollentuna, Sweden) and the purified monoclonal neurofilament H (1 : 400, Catalog Number
RNA was quantified by determining the absorbance at N0142; Sigma, Poole, UK). Primary antibodies were applied
260 nm using a Nanodrop 2000/2000c spectrophotometer for 2 h at room temperature. After rinsing in PBS, secondary
(ThermoScientific, Gothenburg, Sweden). L4, L5, and goat anti-mouse and goat anti-rabbit antibodies Alexa Fluor-
L6 DRGs from rats in each experimental group were 488 and Alexa Fluor-568 (1 : 300; Molecular Probes,
pooled, and 1–5 ng total RNA was converted to cDNA Invitrogen, ThermoFisher Scientific, Stockholm, Sweden)
using a First-Strand cDNA Synthesis Kit (BioRad iScript were applied for 1 h at room temperature in the dark.

Copyright r 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
Apoptotic pathways in DRG neurons Wiberg et al. 3

On some tissue sections, isolectin B4 (IB4)-FITC (1 : 10; Fig. 1


Sigma) was applied. Images were captured using a Nikon
DXM1200 digital camera (Nikon Instruments, Europe,
Amsterdam, The Netherlands).

Statistical analysis
To determine the statistical difference between groups,
one-way analysis of variance complemented by Bonferroni’s
test (GraphPad Prism; GraphPad Software Inc., La Jolla,
California, USA) was used. Statistical significance was set
as a P value less than 0.05. Nonsignificant differences are
reported (P = NS).

Results
Peripheral nerve injury activates caspase-3 in the dorsal
root ganglia
Analyses by quantitative RT-PCR showed that at 7, 14,
and 28 days following nerve transection, the expression
of caspase-3 was significantly (P < 0.05) increased
3.33 ± 0.24-, 3.24 ± 0.11-, and 2.23 ± 0.28-fold, respec-
tively, in comparison with the control (Fig. 1a). Western
blotting showed that nerve injury induced an increased
expression of full-length caspase-3 protein at 7 and
14 days following nerve transection, but we could not
detect the cleaved caspase-3 fragments (Fig. 1b).
However, using immunohistochemistry, we did observe
active caspase-3 in a small number of neurons (Fig. 1c).

Both extrinsic and endoplasmic reticulum-stress


pathways are activated by peripheral nerve injury
Caspase-8, which is a member of the extrinsic apoptotic
pathway, was increased 1.95 ± 0.20-fold (P < 0.05),
3.35 ± 0.24-fold (P < 0.05), and 1.36 ± 0.11-fold (P = NS)
in comparison with the control 7, 14, and 28 days,
respectively, following nerve transection (Fig. 2a).
Western blotting showed that nerve injury induced
caspase-8 protein processing with increased levels of
cleaved intermediate fragment caspase-8 at 14 days fol-
lowing nerve transection, but we could not detect the Expression of caspase-3 in dorsal root ganglia (DRG) after nerve injury.
(a) Quantitative reverse transcription-PCR analysis of caspase-3
active large (17–21 kDa) and small (10–13 kDa) caspase-8 expression in ipsilateral L4, L5, and L6 DRGs at 7, 14, and 28 days after
fragments (Fig. 2b). At 7, 14, and 28 days following nerve peripheral nerve injury. Significant differences are expressed relative to the
control (normal uninjured DRGs); *P < 0.05; n = 5 animals per
transection, the gene expression of caspase-12 was experimental group. (b) Western blot analysis of caspase-3 expression at
increased 3.63 ± 0.28-, 2.72 ± 0.51-, and 3.19 ± 0.70-fold, 7, 14, and 28 days after nerve transection and in control (con) lysates
respectively, in comparison with the control (Fig. 3a; all prepared from pooled DRGs from n = 4 animals per experimental group.
β-Tubulin was used as a loading control. (c) Representative
P < 0.05). The increased expression of caspase-12 was immunohistochemistry of active caspase-3 expression (red) 14 days after
paralleled by an increased expression of calpain, a known peripheral nerve injury (arrow indicates cell with fragmented nucleus).
activator of caspase-12 [8]. At 7, 14, and 28 days following Scale bar = 40 μm. Staining was repeated in three individual animals.
nerve transection, the expression of calpain was increased
2.62 ± 0.17-, 1.91 ± 0.10-, and 1.42 ± 0.05-fold, respec-
tively, in comparison with the control (Fig. 3b; all (Fig. 3c). Western blotting showed that the increased
P < 0.05). In addition to calpain, the increased expression caspase-12 gene expression was associated with activa-
of caspase-12 was paralleled by an increased expression tion of the enzyme. The 38 kDa active fragment of
of caspase-7, which is another known activator of caspase- caspase-12 was increased at 7, 14, and 28 days following
12. At 7, 14, and 28 days following nerve transection, the nerve transection (Fig. 3d). The increased caspase-7 gene
expression of caspase-7 was increased 2.29 ± 0.15-fold expression also correlated with activation of the enzyme,
(P < 0.05), 2.39 ± 0.25-fold (P < 0.05), and 1.63 ± 0.30-fold with increased protein levels observed at 7 and 14 days
(P = NS), respectively, in comparison with the control following nerve transection (Fig. 3d).

Copyright r 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
4 NeuroReport 2018, Vol 00 No 00

Fig. 2 death [6,7]. Several kinds of cellular stress are known to


initiate the intrinsic pathway including for example oxi-
dative stress and depletion of neurotrophic factors [6,7].
The extrinsic pathway is characterized by the presence of
DRs, which are activated by corresponding ligands. So
far, eight different DRs have been identified in humans,
Fas (CD95) being the most described DR. Upon FasL-
mediated activation of Fas, Fas-associated protein with
death domain recruits procaspase-8 and procaspase-10,
forming a death-inducing signaling complex (DISC)
[7,12]. Procaspase-8 is then autoproteolytically cleaved at
the DISC, generating an active protease, which in turn
activates the downstream executioner caspase-3 [7,12].
Type I cells generate high levels of DISC formation and
increased amounts of active caspase-8, which results in
direct activation of the downstream effector caspase-3
[7,12]. Type II cells, however, generate low levels of
DISC formation and require an amplification loop
through caspase-8-mediated processing of Bid to trun-
cated Bid, inducing the release of cytochrome C from
Expression of caspase-8 in dorsal root ganglia (DRG) after nerve injury. the mitochondria with subsequent activation of caspase-3
(a) Quantitative RT-PCR analysis of caspase-8 expression in ipsilateral [7,12].
L4, L5, and L6 DRGs at 7, 14, and 28 days after peripheral nerve injury.
Significant differences are expressed relative to the control (normal In addition, apoptosis can be induced by the ER. The
uninjured DRGs); *P < 0.05; NS, n = 5 animals per experimental group.
(b) Western blot analysis of caspase-8 showing precursor and cleaved ER regulates the intracellular calcium (Ca2 +) home-
intermediate fragment caspase-8 at 7, 14, and 28 days after nerve ostasis in addition to serving as the cellular site of newly
transection and in control (con) lysates prepared from pooled DRGs synthesized secretory and membrane proteins, which
from n = 4 animals per experimental group. β-Tubulin was used as a
loading control. must be properly folded and post-translationally modified
before exit from the organelle. Proper protein folding and
modification requires molecular chaperone proteins as
well as an ER environment conducive for these reactions.
The ER-stress response is mediated by three different When ER luminal conditions are altered or chaperone
ER membrane receptors, activating transcription factor capacity is overwhelmed, so-called ER stress, the cell
6, inositol-requiring enzyme 1, and PERK [9] and we activates signaling cascades to restore a favorable folding
showed that the phosphorylation of PERK was increased environment through activation of three different ER
at 7 and 14 days following nerve transection (Fig. 4a). membrane receptors: activating transcription factor 6,
Analysis of the DRGs by immunohistochemistry indi- inositol-requiring enzyme 1, and PERK [9]. The pro-
cated that phospho-PERK expression was mainly loca-
tective mechanisms include translational attenuation to
lized to IB4-positive neurons in control tissue (Fig. 4b).
limit further accumulation of misfolded proteins, tran-
scriptional activation of genes encoding ER-resident
Discussion chaperones such as BiP/GRP78 and GRP94, and ER-
Current data on the underlying mechanism behind associated degradation, which serves to reduce the stress
injury-induced DRG neuronal cell death are conflicting, and thereby restore the folding capacity by directing
with uncertainties as to which apoptotic pathways are misfolded proteins in the ER back into the cytosol for
involved. Previous research has suggested that apoptosis degradation by the 26s proteasome [9]. If these protec-
in the DRG is mediated by the intrinsic pathway [10,11], tive mechanisms are not sufficient, apoptosis will take
with the role of other apoptotic pathways being largely place through activation of caspase-12 [13].
overlooked.
ER stress is known to be induced in response to spinal
The intrinsic pathway is regulated by the balance cord injury [14]. In this study, we showed that caspase-12
between proapoptotic Bax and antiapoptotic Bcl-2. When mRNA was significantly upregulated in sensory neurons
the equilibrium points towards Bax, the formation of following nerve transection, as well as calpain and
pores take place in the mitochondrial membrane, allow- caspase-7. Furthermore, phosphorylation of PERK was
ing the release of cytochrome C [6,7]. Once released into upregulated, indicating that the ER might be involved in
the cytosol, cytochrome C interacts with apoptotic pro- the injury-induced apoptosis following peripheral nerve
tease activating factor 1 and procaspase-9, forming the injury. Bax and Bak associate not only with mitochondria
apoptosome, resulting in caspase-3 activation and cell membranes but also the ER [15], and mouse fibroblasts

Copyright r 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
Apoptotic pathways in DRG neurons Wiberg et al. 5

Fig. 3

Expression of caspase-12, calpain, and caspase-7 in dorsal root ganglia (DRG) after nerve injury. Quantitative RT-PCR analysis was carried out on
ipsilateral L4, L5, and L6 DRGs to determine the expression of (a) caspase-12, (b) calpain, and (c) caspase-7 at 7, 14, and 28 days after peripheral
nerve injury. Significant differences are expressed relative to the control (normal uninjured DRGs); *P < 0.05; NS, n = 5 animals per experimental
group. (d) Western blot of caspase-12 and caspase-7 expression at 7, 14, and 28 days after peripheral nerve injury and in control (con) lysates
prepared from pooled DRGs from n = 4 animals per experimental group. β-Tubulin was used as a loading control.

lacking both Bax and Bak (Bax− /–/Bak–/–) are resistant to stress-induced apoptosis is still controversial. Some stu-
apoptosis induced by thapsigargin, tunicamycin, and dies show that fibroblasts from caspase-12− / − mice are
brefeldin A [16]. During ER stress, Bax and Bak undergo resistant to apoptosis in response to ER stress [19],
conformational changes and oligomerization in the ER whereas others claim that caspase-12 does not affect the
membrane [16], releasing Ca2 + from the ER into the survival rate, but rather the degree of inflammation [20].
cytoplasm [16]. The increase in the Ca2 + concentration Furthermore, Sanges et al. [21] have proposed that ER
in the cytosol activates m-calpain, which cleaves and stress-induced apoptosis is mediated by calpain, but not
activates procaspase-12 [8]. Activated caspase-12 then by caspases, on the basis of the observation that calpain
cleaves and activates procaspase-9, which in turn acti- inhibitors, but not a pan-caspase inhibitor, block tunicamycin-
vates the downstream caspase cascade including caspase- induced and thapsigargin-induced apoptosis.
3 [13]. Calpain-deficient mouse fibroblasts have reduced
Adult DRGs comprise a heterogenous pool of sensory
ER stress-induced caspase-12 activation and are resistant
to ER stress-associated apoptosis [17]. neurons that can be divided into subpopulations according
to several parameters including neuronal size, neu-
Furthermore, caspase-7 translocates from the cytosol to rochemistry, trophic requirements, and sensory modality
the cytoplasmic side of the ER membrane in response to [22]. It is possible to distinguish between small unmyeli-
ER stress, where it cleaves the prodomain to generate nated afferents and large myelinated afferents; the former
active caspase-12, resulting in increased cell death. In type comprises two populations of cells differentiated by
addition, cotransfection of cells with caspase-12 and the their potential to synthesize neuropeptides: peptidergic
catalytic mutant of caspase-7 not only blocks the cleavage afferents, which have a predominantly nociceptive function
of caspase-12, but it also attenuates cell death [18]. It and identified by their immunoreactivity for calcitonin
should be noted that although caspase-12 is activated gene-related peptide, and nonpeptidergic afferents,
during ER stress, the involvement of caspase-12 in ER which are exclusively nociceptive and identified by their

Copyright r 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
6 NeuroReport 2018, Vol 00 No 00

Fig. 4 The various apoptotic pathways interact. In parallel with


caspase-12 activation, ER stress triggers caspase-8 activation,
resulting in cytochrome C/caspase-9 activation through Bid
processing [24]. We observed an increased expression of
caspase-8 following sciatic nerve transection, indicating that
the extrinsic pathway might also be involved in the injury-
induced apoptosis following peripheral nerve injury.
In addition to our results suggesting the involvement of
caspases 3, 8, and 12 in injury-induced sensory neuron
cell death, Vignesvara et al. [25] observed significantly
elevated levels of cleaved caspase-2, compared with
cleaved caspase-3, following sciatic nerve transection.
Furthermore, siRNA-mediated downregulation of caspase-2
protected 50% of DRG neurons from apoptosis after serum
withdrawal, whereas downregulation of caspase-3 had no
effect on DRG neuron survival.

Conclusion
As adult DRGs comprise a heterogenous pool of sensory
neurons, it can be assumed that several apoptotic path-
ways are involved in the injury-induced cell death. We
speculate that the intrinsic and extrinsic apoptotic path-
ways, together with the ER-stress response, are involved.

Acknowledgements
This research was funded by Umeå University
(a) Western blot for phosphorylated protein kinase R-like endoplasmic (Insamlingsstiftelsen) and Västerbotten County Council.
reticulum kinase (PERK) in control tissue (con) and 7, 14, and 28 days
after peripheral nerve injury. Lysates were prepared from pooled dorsal
root ganglia from n = 4 animals per experimental group. β-Tubulin was Conflicts of interest
used as a loading control. (b) Immunohistochemistry of neurofilament
(NF)-H-positive and isolectin B4 (IB4)-positive control dorsal root There are no conflicts of interest.
ganglion neurons together with phospho-PERK. Arrows show NF-H-
negative neurons that are positive for phospho-PERK. The asterisk
shows an IB4 neuron that is negative for phospho-PERK. Scale References
bar = 40 μm. Staining was repeated in three individual animals. 1 Welin D, Novikova LN, Wiberg M, Kellerth JO, Novikov LN. Survival and
regeneration of cutaneous and muscular afferent neurons after peripheral
nerve injury in adult rats. Exp Brain Res 2008; 186:315–323.
2 Mattsson P, Meijer B, Svensson M. Extensive neuronal cell death following
intracranial transection of the facial nerve in the adult rat. Brain Res Bull
1999; 49:333–341.
binding of IB4. Myelinated afferents innervate peripheral 3 Hu P, McLachlan EM. Selective reactions of cutaneous and muscle afferent
mechanoreceptors and proprioceptors, and are identified neurons to peripheral nerve transection in rats. J Neurosci 2003;
by their binding of the high-molecular-weight neurofila- 23:10559–10567.
4 Tandrup T, Woolf CJ, Coggeshall RE. Delayed loss of small dorsal root
ment H. Specific sensory neuronal subpopulations show ganglion cells after transection of the rat sciatic nerve. J Comp Neurol 2000;
contrasting responses to peripheral nerve injury, as shown 422:172–180.
5 McKay Hart A, Brannstrom T, Wiberg M, Terenghi G. Primary sensory
by the axotomy-induced death of many cutaneous sensory
neurons and satellite cells after peripheral axotomy in the adult rat:
neurons, whereas muscular sensory afferents survive an timecourse of cell death and elimination. Exp Brain Res 2002;
identical insult [1,3,4]. Furthermore, the proportion of 142:308–318.
6 Benn SC, Woolf CJ. Adult neuron survival strategies – slamming on
IB4-positive neurons decreases in axotomized DRGs [23]. the brakes. Nat Rev Neurosci 2004; 5:686–700.
Reid et al. [10] reported that medial gastrocnemius neu- 7 Parrish AB, Freel CD, Kornbluth S. Cellular mechanisms controlling caspase
rons markedly downregulate caspase-3 mRNA in response activation and function. Cold Spring Harb Perspect Biol 2013; 5:a008672.
8 Nakagawa T, Yuan J. Cross-talk between two cysteine protease families.
to axotomy, whereas sural neurons upregulate its gene Activation of caspase-12 by calpain in apoptosis. J Cell Biol 2000;
expression at 1 week after injury, which was hypothesized 150:887–894.
to be attributable to the differing IB4-positive composition 9 Lai E, Teodoro T, Volchuk A. Endoplasmic reticulum stress: signaling the
unfolded protein response. Physiology (Bethesda) 2007; 22:193–201.
of the subpopulations described. In this study, we found 10 Reid AJ, Mantovani C, Shawcross SG, Terenghi G, Wiberg M. Phenotype of
an upregulation of phospho-PERK following axotomy that distinct primary sensory afferent subpopulations and caspase-3 expression
was mainly restricted to IB4-positive neurons, raising the following axotomy. Histochem Cell Biol 2011; 136:71–78.
11 Englezou PC, Esposti MD, Wiberg M, Reid AJ, Terenghi G. Mitochondrial
possibility that the increased susceptibly observed in these involvement in sensory neuronal cell death and survival. Exp Brain Res 2012;
cells is because of activation of ER stress. 221:357–367.

Copyright r 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.
Apoptotic pathways in DRG neurons Wiberg et al. 7

12 Lavrik I, Golks A, Krammer PH. Death receptor signaling. J Cell Sci 2005; 19 Nakagawa T, Zhu H, Morishima N, Li E, Xu J, Yankner BA, et al. Caspase-12
118:265–267. mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by
13 Morishima N, Nakanishi K, Takenouchi H, Shibata T, Yasuhiko Y. An amyloid-beta. Nature 2000; 403:98–103.
endoplasmic reticulum stress-specific caspase cascade in apoptosis. 20 Saleh M, Mathison JC, Wolinski MK, Bensinger SJ, Fitzgerald P, Droin N,
Cytochrome c-independent activation of caspase-9 by caspase-12. J Biol et al. Enhanced bacterial clearance and sepsis resistance in caspase-
Chem 2002; 277:34287–34294. 12-deficient mice. Nature 2006; 440:1064–1068.
14 Penas C, Guzman MS, Verdu E, Fores J, Navarro X, Casas C. Spinal cord 21 Sanges D, Marigo V. Cross-talk between two apoptotic pathways activated
injury induces endoplasmic reticulum stress with different cell-type by endoplasmic reticulum stress: differential contribution of caspase-12
and AIF. Apoptosis 2006; 11:1629–1641.
dependent response. J Neurochem 2007; 102:1242–1255.
22 Lawson SN. Neuropeptides in morphologically and functionally identified
15 Zong WX, Li C, Hatzivassiliou G, Lindsten T, Yu QC, Yuan J, et al. Bax and
primary afferent neurons in dorsal root ganglia: substance P, CGRP and
Bak can localize to the endoplasmic reticulum to initiate apoptosis. J Cell
somatostatin. Prog Brain Res 1995; 104:161–173.
Biol 2003; 162:59–69.
23 Bradbury EJ, Burnstock G, McMahon SB. The expression of P2X3
16 Wei MC, Zong WX, Cheng EH, Lindsten T, Panoutsakopoulou V, Ross AJ,
purinoreceptors in sensory neurons: effects of axotomy and glial-derived
et al. Proapoptotic BAX and BAK: a requisite gateway to mitochondrial neurotrophic factor. Mol Cell Neurosci 1998; 12:256–268.
dysfunction and death. Science 2001; 292:727–730. 24 Jimbo A, Fujita E, Kouroku Y, Ohnishi J, Inohara N, Kuida K, et al. ER stress
17 Tan Y, Dourdin N, Wu C, de Veyra T, Elce JS, Greer PA. Ubiquitous calpains induces caspase-8 activation, stimulating cytochrome C release and
promote caspase-12 and JNK activation during endoplasmic reticulum caspase-9 activation. Exp Cell Res 2003; 283:156–166.
stress-induced apoptosis. J Biol Chem 2006; 281:16016–16024. 25 Vigneswara V, Berry M, Logan A, Ahmed Z. Caspase-2 is upregulated after
18 Rao RV, Hermel E, Castro-Obregon S, del Rio G, Ellerby LM, Ellerby HM, sciatic nerve transection and its inhibition protects dorsal root ganglion
et al. Coupling endoplasmic reticulum stress to the cell death program. neurons from apoptosis after serum withdrawal. PLoS One 2013;
Mechanism of caspase activation. J Biol Chem 2001; 276:33869–33874. 8:e57861.

Copyright r 2018 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Você também pode gostar