Skip to main content

Main menu

  • HOME
  • CONTENT
    • Early Release
    • Featured
    • Current Issue
    • Issue Archive
    • Collections
    • Podcast
  • ALERTS
  • FOR AUTHORS
    • Information for Authors
    • Fees
    • Journal Clubs
    • eLetters
    • Submit
    • Special Collections
  • EDITORIAL BOARD
    • Editorial Board
    • ECR Advisory Board
    • Journal Staff
  • ABOUT
    • Overview
    • Advertise
    • For the Media
    • Rights and Permissions
    • Privacy Policy
    • Feedback
    • Accessibility
  • SUBSCRIBE

User menu

  • Log out
  • Log in
  • My Cart

Search

  • Advanced search
Journal of Neuroscience
  • Log out
  • Log in
  • My Cart
Journal of Neuroscience

Advanced Search

Submit a Manuscript
  • HOME
  • CONTENT
    • Early Release
    • Featured
    • Current Issue
    • Issue Archive
    • Collections
    • Podcast
  • ALERTS
  • FOR AUTHORS
    • Information for Authors
    • Fees
    • Journal Clubs
    • eLetters
    • Submit
    • Special Collections
  • EDITORIAL BOARD
    • Editorial Board
    • ECR Advisory Board
    • Journal Staff
  • ABOUT
    • Overview
    • Advertise
    • For the Media
    • Rights and Permissions
    • Privacy Policy
    • Feedback
    • Accessibility
  • SUBSCRIBE
PreviousNext
Research Articles, Neurobiology of Disease

Elevated TRPV4 Levels Contribute to Endothelial Damage and Scarring in Experimental Spinal Cord Injury

Hemant Kumar, Chang Su Lim, Hyemin Choi, Hari Prasad Joshi, Kyoung-Tae Kim, Yong Ho Kim, Chul-Kyu Park, Hwan Myung Kim and In-Bo Han
Journal of Neuroscience 26 February 2020, 40 (9) 1943-1955; https://doi.org/10.1523/JNEUROSCI.2035-19.2020
Hemant Kumar
1Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea, 13488,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Hemant Kumar
Chang Su Lim
2Department of Energy System Research and Department of Chemistry, Ajou University, Suwon, Gyeonggi-do, Republic of Korea, 16499,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hyemin Choi
1Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea, 13488,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hari Prasad Joshi
1Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea, 13488,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kyoung-Tae Kim
3Department of Neurosurgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea, 41944,
4Department of Neurosurgery, Kyungpook National University Hospital, Daegu, Republic of Korea, 41944, and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Kyoung-Tae Kim
Yong Ho Kim
5Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, Republic of Korea, 13120
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Chul-Kyu Park
5Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, Republic of Korea, 13120
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hwan Myung Kim
2Department of Energy System Research and Department of Chemistry, Ajou University, Suwon, Gyeonggi-do, Republic of Korea, 16499,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
In-Bo Han
1Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea, 13488,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Abstract

Currently, the role of transient receptor potential vanilloid type 4 (TRPV4), a nonselective cation channel in the pathology of spinal cord injury (SCI), is not recognized. Herein, we report the expression and contribution of TRPV4 in the pathology of scarring and endothelial and secondary damage after SCI. TRPV4 expression increased during the inflammatory phase in female rats after SCI and was expressed primarily by cells at endothelial-microglial junctions. Two-photon microscopy of intracellular-free Ca2+ levels revealed a biphasic increase at similar time points after SCI. Expression of TRPV4 at the injury epicenter, but not intracellular-free Ca2+, progressively increases with the severity of the injury. Activation of TRPV4 with specific agonist altered the organization of endothelial cells, affected tight junctions in the hCMEC/D3 BBB cell line in vitro, and increases the scarring in rat spinal cord as well as induced endothelial damage. By contrast, suppression of TRPV4 with a specific antagonist or in female Trpv4 KO mouse attenuated inflammatory cytokines and chemokines, prevented the degradation of tight junction proteins, and preserve blood–spinal cord barrier integrity, thereby attenuate the scarring after SCI. Likewise, secondary damage was reduced, and behavioral outcomes were improved in Trpv4 KO mice after SCI. These results suggest that increased TRPV4 expression disrupts endothelial cell organization during the early inflammatory phase of SCI, resulting in tissue damage, vascular destabilization, blood–spinal cord barrier breakdown, and scarring. Thus, TRPV4 inhibition/knockdown represents a promising therapeutic strategy to stabilize/protect endothelial cells, attenuate nociception and secondary damage, and reduce scarring after SCI.

SIGNIFICANCE STATEMENT TRPV4, a calcium-permeable nonselective cation channel, is widely expressed in both excitable and nonexcitable cells. Spinal cord injury (SCI) majorly caused by trauma/accidents is associated with changes in osmolarity, mechanical injury, and shear stress. After SCI, TRPV4 was increased and were found to be linked with the severity of injury at the epicenter at the time points that were reported to be critical for repair/treatment. Activation of TRPV4 was damaging to endothelial cells that form the blood–spinal cord barrier and thus contributes to scarring (glial and fibrotic). Importantly, inhibition/knockdown of TRPV4 prevented these effects. Thus, the manipulation of TRPV4 signaling might lead to new therapeutic strategies or combinatorial therapies to protect endothelial cells and enhance repair after SCI.

  • blood–spinal cord barrier
  • endothelial cell
  • scarring
  • spinal cord injury
  • TRPV4
  • two-photon microscopy

Introduction

The pathology of spinal cord injury (SCI) is multifaceted, encompassing the initial tissue disruption from the primary trauma and secondary injury progression, involving inflammation, the death of endothelial cells (ECs), neurons, and glia, extracellular matrix remodeling, scarring, and cavitation (Liu et al., 1997; Ling and Liu, 2007). Vascular pathology can be observed minutes after SCI, initiated by the death of ECs, which, along with pericytes, astrocytes, and neurons, comprise the microvasculature under normal conditions (Whetstone et al., 2003; Casella et al., 2006). The death of ECs continues throughout the acute phase of injury (Benton et al., 2008), predominantly at the injury epicenter, causing substantial hemorrhage and disturbance of vascular autoregulation that subsequently contributes to the death of neurons and glial cells (Casella et al., 2002, 2006). Microvessels in the spinal cord support the neural parenchyma not only by supplying blood and oxygen but also by continuously secreting neurotrophins, such as BDNF and angiopoietins, which support regenerating spinal cord tissue (Guo et al., 2008). SCI therapies targeting EC preservation have shown remarkable potential (Fassbender et al., 2011; Kumar et al., 2018c); however, therapeutic endothelial protection/stabilization within penumbral microvasculature remains largely unexplored because of a lack of understanding of the key molecular pathways triggered after SCI.

Most mammalian tissues, including the peripheral nervous system and the CNS (Montell et al., 2002; Ramsey et al., 2006), express transient receptor potential vanilloid type 4 (TRPV4), a calcium-permeable nonselective cation channel (Liedtke et al., 2000; Strotmann et al., 2000) that is currently recognized as a polymodal ionotropic receptor (Kumar et al., 2018a). TRPV4 channels are activated by a wide range of stimuli (Liedtke et al., 2000; Strotmann et al., 2000; Willette et al., 2008; Kumar et al., 2018a), as well as intracellular signaling pathways (Alessandri-Haber et al., 2004, 2006; Grant et al., 2007; Zhao et al., 2014). The abundant presence of ion channels in the plasma membrane of ECs suggests their functional role (Nilius and Droogmans, 2001). Ion channels in ECs control several functions, including intracellular Ca2+ signals, production and release of many vasoactive factors, regulates macromolecules and also involved in controlling intercellular permeability, EC proliferation, and angiogenesis (Nilius and Droogmans, 2001). Reports suggest that increase in TRPV4 activity disintegrated the cell junctions of blood–CSF barrier (Narita et al., 2015). However, TRPV4 expression and its role following SCI have not been characterized.

Here, we show that TRPV4 expression is associated with the severity of injury at the epicenter after SCI. We demonstrate that TRPV4 upregulation and activation disrupt EC organization after SCI and that ECs are better preserved in the absence of TRPV4. Furthermore, the deletion of TRPV4 signaling significantly impacts SCI-induced inflammatory cascades, blood–spinal cord barrier (BSCB) integrity, degree of scarring, and spontaneous locomotor recovery after SCI. Notably, inhibition or deficiency of TRPV4 was beneficial for endothelial factors, such as BDNF, neurotrophins, and angiopoietin, in the CNS after SCI.

Materials and Methods

Subjects and surgical procedures

A total of 211 female Sprague Dawley rats (220–240 g), for this study, were purchased from Orient Bio. Mice heterozygous (B6.129X1-Trpv4<tm1Msz>) for Trpv4 deficiency (Suzuki et al., 2003) were purchased from RIKEN BioResource Centre and intercrossed to generate Trpv4 KO mice. The genotype was examined using PCR with primer sequence as given in Table 1. WT (C57BL/6JJcl) were purchased from Orient Bio served as controls. Thirty-eight female WT and 22 female KO mouse were used in the study. Animals were housed in a facility with 55%–65% humidity at 24 ± 2°C with a 12 h light/dark cycle and free access to food and water. All animal experiments were performed as per the approved guidelines by the Institutional Animal Care and Use Committee of CHA University (IACUC160073) and Principles of laboratory animal care (National Research Council, 2011). Animals were anesthetized for laminectomies exposing the ∼T10 segment of the spinal cord as previously reported (Kumar et al., 2018b). The vertebral column was stabilized and supported by Allis clamps at T8, and T12 spinous processes as previously described (Kumar et al., 2018b,c) (see Fig. 1A). A metal impounder (rats: 20 g/5 min [mild injury], 35 g/5 min [moderate injury], or 50 g/5 min [severe injury]; mice: 20 g/1 min) was then gently applied to T10 dura, resulting in weight compression SCI. Animals were anesthetized for laminectomies exposing the ∼T10 segment of the spinal cord, and a transverse cut was made to create a right hemisection injury. The surgical site was closed after SCI, by suturing the muscle and fascia using silk suture followed by suturing the skin. Povidone-iodine was applied externally to the surgical site, and animals were kept on a heating pad to maintain body temperature until they recovered from anesthesia, and then 5 ml (rats) or 0.5 ml (mouse) of 0.9% sterile saline-injected subcutaneously. Manual bladder expression was performed twice daily (morning and evening) until a bladder reflex was established.

View this table:
  • View inline
  • View popup
Table 1.

Primer sequences for the genes of interest used in the current study

Cell culture, drugs, and treatments

Blood–brain barrier (BBB) hCMEC/D3 cell line (Millipore) and human umbilical vascular endothelial cells (HUVECs; ATCC) were used to understand the biological role of TRPV4 activation or inhibition in ECs. hCMEC/D3 and HUVECs were cultured in fully supplemented endothelial growth medium (PromoCell, Human Centered Science) as per the manufacturer's instructions. GSK1016790A and RN-1734 were purchased from Sigma Millipore. GSK1016790A and RN-1734 were dissolved in DMSO and then used at the functional concentration of 1 and 10 μm, respectively, for in vitro studies. RN-1734 was dissolved in 5% DMSO, 5% N,N-dimethylacetamide and remaining saline and administered intraperitoneally at the dose of 5 mg/kg 1 h after SCI. Rats were killed at 8 h after injury (HPI-8; calcium imaging) or 1 d postinjury (DPI-1; biochemical parameters) after vehicle or RN-1734 treatment. GSK1016790A was first dissolved in DMSO, and then serial dilution was performed to get the final concentration of 50 pmol [0.3% DMSO in aCSF (Tocris Bioscience)]; subsequently, 10 μl was injected at ∼T10 level of spinal cord with the help of Legato 130 Syringe Pump (KD Scientific).

Behavioral assessment

Hindlimb locomotor score.

Hindlimb locomotor function was evaluated using the open-field Basso Mouse Scale (BMS) locomotor test (Basso et al., 2006) on 1, 3, 7, 14, 21, and 28 d following injury in WT (n = 10) and TRPV4 KO (n = 10) mice. The scoring ranged from 0 points (no ankle movement) to 9 points (complete functional recovery). The animal's hindlimb motor functions were evaluated by two experienced investigators who were blinded to treatment group.

Test for nociception.

Nociception in mice was assessed using the hotplate method at pre-SCI (basal), 1, 3, 7, 14, 21, and 28 d after SCI. Briefly, mice (n = 6/group) were placed on the surface of the hotplate (25.4 × 25.4 cm; Ugo Basile) heated to 54°C, which was surrounded by a transparent Plexiglas chamber (see Fig. 6A). The latency to respond was measured with either a hindpaw lick or flick, with a cutoff threshold of 20 s to prevent tissue injury. Mice were removed immediately from the hotplate after a response. A total of three readings were used to determine the average reaction time.

qRT-PCR

qRT-PCR was performed at respective time points using a SYBR Green Master Mix, and the mRNA detection was analyzed using an ABI StepOne Real-time PCR System (Applied Biosystems) (Kumar et al., 2018c). Primer sequences for the genes of interest used in the current study were as given in Table 1: Typical profile times were the initial step, 95°C for 10 min followed by a second step at 95°C for 15 s, and 60°C for 30 s for 40 cycles with a melting curve analysis. The target mRNA level was normalized with the level of the GAPDH and compared with the control. Data were analyzed using the ΔΔCT method.

Western blot analysis

hCMEC/D3 cells were collected after 24 h of drug treatment. Spinal cord tissues were collected at DPI-1 and DPI-28 and washed with PBS, placed at 4°C, and homogenized in lysis buffer (PRO-PREP, iNtRON Biotechnology) and centrifuged at 14,000 rpm at 4°C for 15 min. The supernatant was collected for determination of protein concentration using Bio-Rad DC Protein Assay. Protein concentration was determined by VersaMax microplate reader. Equal amounts of protein (40 μg) were separated electrophoretically by 10% SDS-PAGE electrophoresis, and the resolved proteins were transferred to PVDF membranes (#162-0177, Bio-Rad). The membranes were then incubated for 1 h with 5% nonfat skim milk prepared in TBS buffer to block nonspecific binding. The membranes were then incubated overnight in cold room with primary antibodies as given in Table 2. After 1 h incubation at room temperature with corresponding secondary antibodies, the blots were visualized with enhanced chemiluminescence (GE Healthcare), using the LAS 4000 biomolecular imager (GE Healthcare). The immunoblots were quantified using ImageJ software (Fiji).

View this table:
  • View inline
  • View popup
Table 2.

Primary antibody information used for immunoblot analysis

Immunohistochemistry and Immunofluorescence

After SCI at T10, animals were anesthetized and perfused with 0.9% saline, followed by 4% PFA for tissue fixation at several time points as per the time points and treatments. The spinal cord at compression site was retrieved and immersed in 4% PFA for 1 d, and then fixed in paraffin, sectioned at 5 μm, dewaxed, and stained with primary antibodies overnight at 4°C followed by secondary antibody as given in Tables 3 and 4. Following PBS washing, DAPI (1:500) was incubated for 10 min. Sections were mounted in ACRYMOUNT mounting media (StatLab) and examined using a fluorescence microscope (Carl Zeiss or Leica Microsystems). Image quantification for immunofluorescence signals was performed using Zen 3.0 [Blue edition (Carl Zeiss)]. Equal area (nm2) was selected, channel intensities were measured, and fluorescence intensity mean value (IMV) was obtained. The fluorescence IMV was plotted as mean ± SEM in GraphPad Prism (version 5.01, GraphPad Software).

View this table:
  • View inline
  • View popup
Table 3.

Primary antibody information used for immunohistochemistry analysis

View this table:
  • View inline
  • View popup
Table 4.

Secondary antibody information used for immunohistochemistry analysis

Immunocytochemistry

For immunofluorescence microscopy, third passage hCMEC/D3 BBB cell line and HUVECs (0.5 × 105 cells/well) were cultured on a sterile rat tail collagen-I (Sigma Millipore)-coated coverslip in 24-well plates. Immunocytochemistry was performed as per previously published protocol (Kumar et al., 2018b). Cells were incubated overnight at 4°C with antibodies directed against TRPV4 (1:100, Alomone Labs) and CD-31 (1:100, Abcam). Secondary antibodies (1:500) were goat anti-rabbit AlexaFluor-488 and goat anti-mouse AlexaFluor-488 (Abcam). Following DAPI incubation for 10 min, coverslips were mounted and examined using a fluorescence microscope (Carl Zeiss).

Calcium imaging using two-photon fluorescence microscopy

We evaluated the [Ca2+]i inside the live rat or mouse spinal cord tissue after the injury as per the previously reported method (Kim et al., 2017). Briefly, a thin sectional tissue slice was extracted from the injured area following SCI at 3 and 8 HPI and at 1, 3, 5, 7, 14, 21, and 28 DPI for effective two-photon microscopy (TPM) imaging. Consequently, we incubated the spinal cord sectional slice with 10 μm Ca2+ sensing unit SCa1-IREF for 40 min at 37°C, and ratiometric TPM images were acquired at depths of 90–210 μm for visualization of the overall Ca2+ distribution (Kim et al., 2017). The TPM images of SCa1-IREF-labeled tissues were acquired with spectral confocal and multiphoton microscopes (Leica Microsystems, TCS SP8 MP) with × 10 dry and × 40 oil objectives, numerical aperture (NA) = 0.40 and 1.30 as previously described (Kim et al., 2017). Ratiometric image processing and analysis were performed using MetaMorph software (Molecular Devices).

Evaluation of blood–spinal cord barrier permeability

To establish the role of TRPV4 in BSCB disruption, we examined samples from sham, injured WT, and injured TRPV4-KO mouse prepared DPI-1. The BSCB permeability was investigated with Evans blue dye extravasation according to a previous report (Kumar et al., 2018b). Briefly, at DPI-1 after SCI, 500 μl of 2% Evans blue dye (Sigma Millipore) solution prepared in saline and administered intraperitoneally. Mouse were anesthetized 3 h after Evans blue dye injection and killed by intracardiac perfusion with saline. Spinal cord segment was collected and homogenized in 50% trichloroacetic acid solution. Homogenate samples were centrifuged at 10,000 × g for 10 min, and supernatant was collected. Fluorescence intensity of Evans blue was quantified using a spectrophotometer at excitation wavelength of 620 nm and emission wavelength of 680 nm as previously described (Kumar et al., 2018b).

TUNEL assay

Paraffin sections from TRPV4 KO and WT mouse were examined for apoptotic cells at DPI-1 using an in situ death detection kit (Roche Diagnostics) as per the manufacturer's information. The images were examined using an Olympus microscope (U-TVO.63XC).

Statistical analyses

Statistical analysis was performed using GraphPad Prism (version 5.01, GraphPad Software). Data are presented as mean ± SEM from the indicated number of experiments. The in vivo PCR data were analyzed using one-way ANOVA, followed by Tukey's test. The BMS scores and hotplate data were analyzed statistically with two-way ANOVA followed by the Bonferroni test. p values < 0.05 were considered statistically significant. The respective statistical analysis used to analyze the data is mentioned in the figure legends.

Results

TRPV4 expression increases during the early phase of SCI

We first characterized the time course of TRPV4 expression in rats at the epicenter of the SCI at 3 and 8 HPI and at 1, 3, 5, 7, 14, 21, and 28 DPI (35 g for 5 min) (Fig. 1A). TRPV4 expression was low in the uninjured spinal cord (sham condition), localized mainly to ECs, blood vessels, and neurons, and substantially increased during the early phase after SCI (Fig. 1C,F; Fig. 1-1A, ). The integrity of the vascular endothelium was assessed by immunostaining for rat EC antigen (RECA) (Fig. 1D; Fig. 1-1B) and angiopoietin-1 [(ANG-1), primarily expressed by pericytes] (Fig. 1-1E,F). Neurofilament (NF), a marker of the neuronal cytoskeleton (Fig. 1D; Fig. 1-1B,E,F) was used to evaluate the neuronal damage after injury. ECs were lost in the injury epicenter as early as HPI-3, with some recovery by DPI-5, whereas NF immunoreactivity gradually declined with time after injury (Fig. 1D; Fig. 1-1B,D,E,F). Intracellular calcium concentrations ([Ca2+]i) were quantitated in situ at the same time points by using TPM (Fig. 1B), which revealed a biphasic response to SCI (Fig. 1E,G; Fig. 1-1C). SCI induced a secondary injury cascade via the production of inflammatory mediators, such as IL-6. IL-6, as assessed by qRT-PCR, showed a transient increase at early time points from HPI-3 to DPI-1 after injury (Fig. 1H). qRT-PCR analysis showed the altered expression of protein kinase C and casein kinase substrate in neurons 3 (Pacsin-3), a member of the family of proteins involved in synaptic vesicular membrane trafficking that also strongly inhibits TRPV4 expression (D'hoedt et al., 2008) (Fig. 1I).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

TRPV4 expression increased during the inflammatory/acute phase of SCI. A, Schematic showing SCI instrument and method. B, Schematic illustration showing TPM imaging after SCI. Representative immunohistochemistry images performed on longitudinal sections at epicenter of the damage for TRPV4 (C), and NF and RECA-1 (D) at 3 h, 8 h, and 1, 3, 5, and 7 d after moderate static compression (35 g/5 min) SCI (Carl Zeiss microscope, 3 fields/slide, n = 3/time point). SCI rat sectional slice labeled with 10 μm SCa1-IREF for 40 min. In situ Ca2+ levels were determined in the transverse spinal cord using TPM at similar time points after SCI at the epicenter of the damage. E, Two-photon excited fluorescence was collected using 750 nm excitation and emission windows at 400–430 nm (Ch1) and 500–600 nm (Ch2). Total RNA was prepared from the epicenter of the damage collected 3 h, 8 h, and 1, 3, 5, 7, 14, 21, and 28 d after SCI to determine the expression of TRPV4 (F), IL-6 (H), and Pacsin-3 (I) (n = 4–6/time point, performed in triplicates). G, Quantification of in situ Ca2+ levels. GAPDH was used as internal controls for qRT-PCR. Data are mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 compared with sham group. $p < 0.05, $$$p < 0.001 compared with HPI-3. #p < 0.05 compared with HPI-8. @p < 0.05, @@p < 0.01, @@@p < 0.001 compared with DPI-1. ∧p < 0.05, ∧∧p < 0.01 compared with DPI-3. np < 0.05, nnp < 0.01 compared with DPI-5. gp < 0.05, gggp < 0.01 compared with DPI-7. xp < 0.05, xxxp < 0.01 compared with DPI-21 by one-way ANOVA Tukey's post test.

Figure 1-1

TRPV4 expression increased during the acute phase of SCI Representative images of immunohistochemistry performed on longitudinal sections at epicenter of the damage for Transient receptor potential cation channel subfamily V member 4 (TRPV4) (A), and Neurofilaments (NF) and rat endothelial cell antigen (RECA-1) (B) at 14, 21 and 28 days after moderate static compression (35 G/5min) SCI [3 fields/slide, n = 3/group]. Bar charts show the fluorescence intensity mean value (IMV) for RECA and NF (D) as per randomly selected field area at the injury epicenter (3 fields/slide, n = 2-3/group). In-situ Ca2+ levels were determined in the transverse spinal cord using TPM at similar time points after SCI at the epicenter of the damage (C). Western blots and quantification of angiopoietin-1 (ANG-1), and Neurofilament (NF) expression at 8 h and 1, 3, 5, 7, 14, 21, and 28 days after injury (E-G). Actin was used as internal controls for western blot [n = 2/group]. Data represent means ± S.E.M. *p < 0.05, ***p < 0.001, ***p < 0.001 vs. sham group (D, F and G). Download Figure 1-1, TIF file

Injury-dependent TRPV4 expression at the epicenter of SCI

Next, we also investigated whether TRPV4 followed an injury-dependent expression. Rats were subjected to mild (20 g/5 min), moderate (35 g/5 min), and severe (50 g/5 min) SCI (Fig. 2A–C). TRPV4 expression increased with the severity of the injury at the epicenter area compared with the sham group (Fig. 2D,I; Fig. 2-1A). The expression of TRPV4 was also increased at the rostral area in mild and severe SCI (Fig. 2D). However, the increase in [Ca2+]i measured by TPM at HPI-8 did not correspond to the severity of the injury (Fig. 2J,H). Pacsin-3 expression was declined with an increase in the injury (Fig. 2F). Assessments of inflammation (IL-6 [Fig. 2E]; HO-1, ANG-2, and TNF-α [Fig. 2-1B,D,I], BSCB integrity, endothelial and vascular damage (occludin [Fig. 2G]; RECA [Fig. 2L]; neuropilin, ANG-2, and ANG-1[Fig. 2-1C,D,J], and neuronal damage (NF [Fig. 2L]), were congruent with the injury severity. TRPV4 immunoreactivity assessed at DPI-1 colocalized with that for the microglial marker Iba-1 and for endothelial-specific marker CD-31, indicating that ECs and microglia express TRPV4 after SCI (Fig. 2K). Furthermore, the expression of TRPV4, IL-6, and Pacsin-3 was altered by hemisection of the spinal cord; however, the increase in TRPV4 was to a lesser extent than that with compression SCI (Fig. 2M–O). Together, these results suggest that TRPV4 expression and related parameters are associated with the severity of compression injury.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

TRPV4 expression is linked with injury severity and inflammation. Schematic showing method for injury-dependent expression and impounder of different weight (A,B). H&E staining was performed on the longitudinal section after mild (20 g/5 min), moderate (35 g/5 min), and severe (50 g/5 min) injury (C). Total RNA was prepared from spinal cord tissues at the rostral, epicenter, and caudal of the damage collected at 1 d after SCI (mild, moderate, and severe compression) to determine the expression of TRPV4 (D), IL-6 (E), Pacsin3 (F), and occludin (G). RNA expression was determined at the epicenter at 1 d after SCI (mild, moderate, and severe) (n = 2 or 3/group performed in triplicates). TRPV4 (I) and NF and RECA-1 (L) IHC was performed after mild (20 g), moderate (35 g), and severe (50 g) injury (Carl Zeiss microscope, 3 fields/slide, n = 3/time point). In situ Ca2+ levels were determined using TPM at 8 h after SCI (mild, moderate, and severe) at the epicenter of the damage (J). Quantification of in situ Ca2+ levels (H). Colocalization of TRPV4 with CD-31, an endothelial marker, and Iba-1, a microglial marker (K). Total RNA was prepared from spinal cord tissues after spinal cord hemisection to determine the expression of TRPV4 (M), IL-6 (N), and Pacisn3 (O) (n = 4 or 5/group). GAPDH was used as internal controls for qRT-PCR. Data are mean ± SEM performed in triplicates. *p < 0.05, **p < 0.01, ***p < 0.001 compared with sham group. $p < 0.05, $$p < 0.01, $$$p < 0.001 compared with 20 g injury group. #p < 0.05, ##p < 0.01, ###p < 0.001 compared with 35 g injury group by one-way ANOVA Tukey's post test.

Figure 2-1

TRPV4 expression followed injury dependent pattern Representative images of immunohistochemistry performed on transverse sections at -5 mm, 0 mm, and +5mm of the damage for Transient receptor potential cation channel subfamily V member 4 (TRPV4) at 1 day after mild (20G/5min), moderate (35G/5min), and severe (50G/5min) static compression [n =3/group] (A). Total RNA was prepared from spinal cord tissues at the epicenter of the damage collected at 1 day after SCI (mild, moderate and severe compression) to determine the expression of HO-1 (B), Neuropilin-1 (C), Angiopoietin-2 (D), Nucleophosmin (E), Arachidonate 15 lipoxygenase (F), Aquaporin-9 (G), KCJN 10 (H), TNF-alpha (I) and Angiopoietin-2 (J) [n = 2-3/group performed in triplicates]. Data represent means ± S.E.M. *p < 0.05, ***p < 0.001, ***p < 0.001 vs. sham group (B-J). Download Figure 2-1, TIF file

Activation of TRPV4 promotes endothelial damage, whereas inhibition is protective

To evaluate the effect of TRPV4 activation on ECs and the BSCB, we incubated hCMEC/D3 BBB cells, which have basal expression of TRPV4, for 24 h with a TRPV4 agonist (1 μm GSK1016790A [GSK]) or antagonist (10 μm RN-1734), which results in increased or decreased TRPV4 expression, respectively (Fig. 3A,B). The CD-31 immunoreactivity to assess the integrity/status of the ECs was reduced by TRPV4 activation; this reduction was mitigated by pretreatment with the antagonist (Fig. 3A,C). Accordingly, TRPV4 agonist or antagonist results in increased or decreased TRPV4 expression, respectively, in HUVECs (Fig. 3D,E). Similar to hCMEC/D3 BBB cells, endothelial integrity/status, as measured by von Willebrand factor (vWF) immunoreactivity, was altered by TRPV4 activation, and this was prevented by the TRPV4 antagonist (Fig. 3D,F).The expression of TJ markers zonula occludens-1 (ZO-1), ZO-2, and claudin-1 was attenuated by GSK (1 μm), with a prominent effect observed for ZO-2 and claudin-1; however, the TRPV4 antagonist RN-1734 (10 μm) or RN-1734 treatment before GSK either maintained or increased the expression of the TJ markers (Fig. 3-1A). To assess the effect of TRPV4 activation in vivo, GSK (50 pm) was injected directly into the spinal cords of rats. We examined samples from uninjured sham and GSK-treated animals after 28 d as well as from vehicle and GSK-treated animals at DPI-28. SCI causes centralized fibrotic scars surrounded by a reactive glial scar at the epicenter. Damaged vessels immunoreactive for laminin, a component of the basal lamina, were readily identified at DPI-28 (Fig. 3G). A single injection of GSK changed/damaged the morphology of laminin structures (J- or T-shaped under normal conditions) and RECA-stained vessels (Fig. 3G,J). The basal lamina also comprises collagen IV, and the normal uniform distribution was altered by GSK injection; a dense collagen IV meshwork was observed at DPI-28 in animals treated with vehicle or GSK (Fig. 3-1N,O, ). Immunoreactivity for ANG-1, a CNS endothelium neurotrophin, was decreased after GSK treatment (Fig. 3-1N,O).

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

TRPV4 activation causes endothelial remodeling/damage, whereas TRPV4 inhibition protects SCI-induced endothelial damage. TRPV4 and CD-31 (A) immunocytochemistry was performed on fixed hCMEC/D3 BBB cell line as described in Materials and Methods. Quantification of TRPV4 and CD-31 fluorescence (B,C). TRPV4 and vWF immunocytochemistry was performed on fixed HUVECs (D) and its quantification (E,F). GSK1016790A (50 pmol, 10 μl) was injected into the spinal cord of the rats as mentioned in Materials and Methods. Samples from sham, GSK1016790A, vehicle (injury), or GSK1016790A (injury) were prepared 28 d after injection/injury. Representative images of laminin (magenta) and RECA-1 (G; green), GFAP (H; green), Iba-1 (H; red), and NFs (I; red). J–L, Bar charts represent the fluorescence IMV for corresponding protein as per randomly selected field area at the injury epicenter (3 fields/slide, n = 3/group). Samples from sham or injured untreated (injury) or after RN-1734 (5 mg/kg) treatment were prepared 1 d after moderate injury (35 g/5 min). Representative transverse section of TRPV4 (M) and ZO-1 (N) (3 fields/slide, n = 3) and fluorescence IMV for corresponding protein (P,Q). Total RNA from sham, vehicle (injury), or RN-1734-treated (5 mg/kg, i.p.) samples was prepared DPI-1 after injury. RT-PCR results are showing relative expression levels of TRPV4 (S), Pacsin3 (T), inducible nitric oxide synthase (U), IL-6 (V), and occludin (W). In situ Ca2+ levels were determined in the transverse spinal cord using TPM at 8 h after SCI at the epicenter of the damage (O). Quantification of in situ Ca2+ levels (R). Data are mean ± SEM (n = 2 or 3/group performed in triplicates). *p < 0.05, **p < 0.01, compared with sham group. $p < 0.05, $$p < 0.01, $$$p < 0.001 compared with GSK group. #p < 0.05 (B,C,E). #p < 0.05, ##p < 0.01, ###p < 0.001 compared with sham group. *p < 0.05, **p < 0.01, compared with injury group (P–W). *p < 0.05, **p < 0.01, ***p < 0.001 compared with sham group. $p < 0.05, $$p < 0.01, $$$p < 0.001 compared with GSK group (J–L) by one-way ANOVA Tukey's post test.

Figure 3-1

TRPV4 activation cause endothelial remodeling/damage, whereas TRPV4 inhibition protects SCI-induced endothelial damage Western blot analysis was performed in hCMEC/D3 BBB cell line collected 24 h after treatment with GSK1016790A (1μM), RN-1734 (10 μM) and RN-1734 (10 μM) + GSK1016790A (1μM) [hCMEC/D3 BBB cell line exposed only to DMSO served as the control] (A). Western blots of zonula occludens (ZO)-1, ZO-2, Claudin-1, CD2-associated protein, Afadin), and TRPV4 (A). Actin was used as internal controls for western blot [n = 2-3/group]. Quantification of immunoblot for ZO-1 (B), ZO-2 (C), Claudin-1 (D), CD2AP (E), Afadin (F), and TRPV4 (G) was performed using ImageJ. GSK1016790A (50 pmol, 10 μl) was injected into the spinal cord of the rats as mentioned in Methods sections. Samples from sham, GSK1016790A, vehicle (Artificial CSF containing 0.3% DMSO; injury) - or GSK1016790A (injury) were prepared 28 days after injection/injury. Representative images of Collagen IV (green) and angiopoietin-1 (ANG-1; magenta) and merge (N). Bar charts show the fluorescence intensity mean value (IMV) for Collagen IV and ANG-1 (O) at the injury epicenter (n = 3/group). Representative images of H & E staining at DPI-28 after injury/injection (P). Samples from sham, vehicle (injury) - or RN-1734-treated (5mg/kg, i.p.) were prepared DPI-1 after injury. Western blots and quantification of TRPV4 and IL-6 expression at 1day after injury (H-J) [n = 2-3/group]. Representative merges images (transverse) for nitrotyrosine and CD-86 at DPI-1 and their quantification (K-M). Data represent means ± S.E.M. *p < 0.05 vs. control group, $p < 0.05 vs. GSK group (B-G). ###p < 0.001 compared with sham group, *p < 0.05, ***p < 0.001 vs. injury group (I-M). ***p < 0.001 vs. sham group, ### p < 0.001 vs. GSK group, @p < 0.05 vs DPI-28 (Vehicle) group (O). Download Figure 3-1, TIF file

We also evaluated the effect of TRPV4 activation alone or with injury at day 28. Interestingly, GSK injections only produced an increase in microglial cells (as determined by Iba-1 immunoreactivity) but not in reactive astrocytes (as determined by GFAP immunoreactivity) (Fig. 3H,K). Immunoreactivity for the axonal marker NF revealed significant loss with GSK injection and extensive damage in injured spinal cords compared with that in the sham group (Fig. 3I,L). Additionally, blood infiltration following BSCB disruption after SCI initiates a secondary injury cascade via the production of inflammatory mediators, such as TNF-α, IL-6, and inducible nitric oxide synthase. As the activation of TRPV4 appeared to damage ECs, we examined the effect of TRPV4 inhibition on BSCB integrity, inflammation, and Ca2+ levels during the early phase after SCI, when there is peak TRPV4 expression. Samples from sham, injured vehicle-treated, and injured RN-1734-treated (5 mg/kg, i.p.) animals were analyzed at DPI-1. Treatment with RN-1734 significantly inhibited TRPV4 expression attenuated SCI-induced inflammatory cytokines and prevented the loss of the TJ proteins occludin and ZO-1 after injury (Fig. 3M,N,P,Q,S–W; Fig. 3-1H–M). In addition, treatment with RN-1734 significantly reduced the SCI-induced calcium release as determined by Ca2+ imaging at HPI-8 (Fig. 3O,R).

TRPV4 KO mice have less endothelial damage and inflammation

On the basis of the above-described results, we hypothesized that the ablation of TRPV4 signaling would protect ECs and preserve BSCB integrity. To test this, we induced SCI (20 g/1 min) at ∼T10 in TRPV4 KO and WT mice at DPI-1 (Fig. 4A). Evans blue extravasations consistent with an increase in barrier permeability were observed in WT mice but not in TRPV4 KOs (Fig. 4B,C). The loss of TJ proteins, such as occludin, ZO-1, ZO-2, and claudin-1, did not occur in TRPV4 KO mice (Fig. 4D,L,Q; Fig. 4-1A,C–F), and ECs were preserved, as determined by higher CD-31 levels (Fig. 4-1A,G), accompanied by an attenuation of the increase in VEGF expression after injury (Fig. 4E). The ∼100 kDa band for TRPV4 was barely detectable in the KO mice compared with the WT after injury (Fig. 4-1A,B). Proinflammatory cytokine and chemokine production increased in WT mice after SCI compared with that in sham controls, and this was significantly attenuated in TRPV4 KO mice (Fig. 4F–K).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Reduced endothelial damage and inflammation after SCI in TRPV4 KO mice. Representative images of H&E staining at DPI-1 after SCI (A). Representative whole spinal cords show Evans blue dye extravasation of the spinal cord 1 d after SCI (B) and its quantification (C). Total RNA from sham (black bar), wild (red bar), or TRPV4 KO (blue bar) samples was prepared DPI-1 after the injury as described in Materials and Methods. RT-PCR results show relative expression levels of occludin (D), VEGF (E), IL-1β (F), IL-6 (G), TNF-α (H), macrophage-1 antigen (Mac-1) (I), chemokine (C-C motif) ligand 2 (CCL-2) (J), and chemokine (C-C motif) ligand 3 (CCL-3) (K) after injury (n = 2 or 3/group performed in triplicates). GAPDH was used as internal controls for qRT-PCR. TUNEL assay was performed at DPI-1 (L). Quantification of TUNEL-positive cells (M). SCI-induced mouse sectional slice labeled with 10 μm SCa1-IREF for 40 min. In situ Ca2+ levels were determined in the longitudinal spinal cord using TPM at 8 h after SCI at the epicenter of the damage. Two-photon excited fluorescence was collected using 750 nm excitation and emission windows at 400–430 nm (Ch1) and 500–600 nm and its quantification (Ch2) (N,O). Representative images for ZO-1 (magenta) and occludin (Q; green) and fluorescence IMV for corresponding protein (P). ###p < 0.001 compared with sham group. *p < 0.05, **p < 0.01, ***p < 0.001 compared with WT-injury group (D–K,O) by one-way ANOVA Tukey's post test. *p < 0.05, **p < 0.01, ***p < 0.001 vs WT-injury group (C,M,P) by unpaired t test.

Figure 4-1

Reduced endothelial damage and inflammation after spinal cord injury in TRPV4 KO mice Representative Western blots of TRPV4, zonula occludens (ZO)-1, ZO-2, occludin, claudin-1, CD-31, p-38, AKT, JNK, ERK, and caspase-3 expression at 1day after injury (A). Actin was used as internal controls for western blot. Quantification of immunoblot for TRPV4 (B), ZO-1 (C), ZO-2 (D), Occludin (E), Claudin-1 (F), CD-31 (G), p-38 (H), AKT (I), JNK (J), ERK (K) and Caspase-3 (L) was performed using ImageJ. Data represent means ±S.E.M. *p < 0.05, **p < 0.01, ***p < 0.001 vs. WT-Injury group. KO vs. WT data was analyzed using the unpaired t-test. Download Figure 4-1, TIF file

We performed TUNEL assays to assess apoptosis after SCI. At DPI-1, apoptotic cells were present at the epicenter in both WT and KO mice; however, the percentages of TUNEL-positive cells and the amount of caspase-3 were much lower in TRPV4 KO mice than in WT mice (Fig. 4L,M; Fig. 4-1A,L). ERK, JNK, and AKT expression was not significantly different between TRPV4 KO and WT mice, whereas KO mice showed a reduction in p38 expression (Fig. 4-1A,H–K). TPM imaging at HPI-8 revealed that SCI-induced [Ca2+]i was inhibited in TRPV4 KO mice (Fig. 4N,O).

Reduced scarring in TRPV4 KO mice after SCI

To further assess the endothelial protective effects of TRPV4 ablation on scarring, we assessed scarring (glial and fibrotic) 28 d after SCI. Whereas the lesions of WT animals contained large amounts of laminin and collagen IV, hallmarks of the fibrotic scars thought to hinder axon regeneration, the amounts in lesions of TRPV4 KO mice were much lower (Fig. 5A,B,G,I). The expression of GFAP indicative of glial formation was higher at DPI-28 in WT than in TRPV4 KO mice, whereas the expression of NF, a marker for neuronal cytoskeleton, was higher at DPI-28 in TRPV4 KO than in WT mice, suggesting that KO mice had attenuated glial scarring and better neuronal protection (Fig. 5C,K,M,N,Q). Similarly, immunostaining for Iba-1 to detect resident microglia and infiltrating monocyte-derived macrophages showed significantly reduced expression in TRPV4 KO mice compared with that in WT mice (Fig. 5D,H,M,O). By contrast, the expression of CD-206, a marker for M2 macrophages, was increased in TRPV4 KO mice (Fig. 5E,J). Immunoreactivity for TGF-β, a key mediator of fibrotic scarring, and CD-68, a marker for activated macrophages, was significantly lower in TRPV4 KO mice than in WT mice (Fig. 5F,L). The expression of connexin-43, an abundant gap junction protein in the CNS, was markedly higher in WT mice than in TRPV4 KO mice, whereas CD-31, which makes up a large portion of EC intercellular junctions, was higher in TRPV4 KO mice (Fig. 5M,P,R).

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

TRPV4 KO mice exhibit less fibrotic and glial scarring after SCI. Samples from WT (injury) or TRPV4 KO (injury) were prepared at DPI-28 as described in Materials and Methods. Representative merges images (longitudinal) for Collagen IV (A; white), laminin (B; magenta), GFAP (C; green), NF (C; magenta), Iba-1 (D; magenta), CD-206 (E; green), CD-68 (F, green), and TGF-β1 (F; magenta) at DPI-28. Bar charts represent the fluorescence IMV for Collagen IV (G), Iba-1 (H), laminin (I), CD-206 (J), GFAP and NF (K), and TGF-β1 and CD-68 (L) as per randomly selected field area at the injury epicenter (3 fields/slide, n = 3/group). M, Western blots of NFs, Iba-1, Connexin-43, GFAP, and CD-31 expression at 28 d after injury. Quantification of immunoblot for NF (N), Iba-1 (O), Connexin-43 (P), GFAP (Q), and CD-31 (R) was performed using ImageJ. Actin was used as internal controls for Western blot (n = 2 or 3/group). Data are mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 vs WT-injury group by unpaired t test.

TRPV4 KO mice show improved functional recovery and neuroprotection

To evaluate the functional consequences of TRPV4 ablation, we assessed gross locomotor function in TRPV4 KO and WT mice using the BMS to assess open field hindlimb locomotion (Basso et al., 2006). TRPV4 KO mouse spinal cord was better preserved compared with WT as determined by H&E staining (Fig. 6A). Mice developed paraplegia after SCI, corresponding to a low BMS score at DPI-1-3, but demonstrated evidence of modest improvement as early as DPI-7 (Fig. 6B). The BMS scores over 4 weeks ranged from 0.30 ± 0.48 to 4.7 ± 2.0 for WT mice and from 0.50 ± 0.71 to 7.00 ± 2.00 for TRPV4 KO mice, with TRPV4 ablation associated with faster recovery and better motor coordination in comparison with that for WT (Fig. 6B). The performance of sham control mice remained unchanged throughout the testing period (data not shown). As nociception is also affected after SCI, we evaluated the effect of TRPV4 deletion on SCI-induced pain using the hotplate test (Fig. 6C). After SCI, animals exhibited thermal hypersensitivity (decrease in reaction time) on days 1, 3, 7, and 14 but not at 21 and 28 d (Fig. 6D). This hypersensitivity was significantly attenuated at DPI-1, DPI-3, and DPI-7 in TRPV4 KO mice. Immunoreactivity for ANG-1, BDNF, and neurotrophin 3 (NT-3) neurotrophic factors, which are primary mediators of axonal and neuronal plasticity and regeneration after SCI, was also greater in TRPV4 KO mice than in WT mice after SCI (Fig. 6E,F). We also assessed the effects of TRPV4 deletion on neuroprotection and angiogenesis after SCI. Immunohistochemistry of microvessels in spinal cord sections revealed that expression of vWF (Fig. 6G) and neural/glial antigen2 (NG-2) and α-smooth muscle actin (α-SMA) (Fig. 6H) was higher in TRPV4 KO mice after injury than in WT mice. Accordingly, immunoreactivity for growth cones was more evident in axons in TRPV4 KO mice than in WT mice (Fig. 6J), which was accompanied by greater immunoreactivity for neuronal markers TuJ1 and NeuN, indicating better neuronal protection in TRPV4 KO mice (Fig. 6I,J). These results suggest that abrogation of TRPV4 signaling is neuroprotective during SCI.

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

TRPV4 KO mice display reduced hyperalgesia, improved functional recovery, enhanced neuroprotection, and endothelial preservation after SCI. A, Representative images of H&E staining at DPI-28 after SCI. B, Functional recovery was assessed in open-field testing by using the 9-point BMS locomotor test at 1, 3, 7, 14, 21, and 28 d after SCI (n = 10/group). C, Nociception was evaluated using hotplate test. D, SCI-induced hypersensitivity (decrease in reaction latency time) was assessed at pre-SCI (basal), 1, 3, 7, 14, 21, and 28 d after SCI (n = 6/group). Samples from WT (injury) or TRPV4 KO (injury) were prepared at DPI-28 as described in Materials and Methods. Representative sections of angiopoietin-1 (ANG-1, white), NT-3 (red), and BDNF (green) in WT (E) and KO (F). Representative section of vWF (G; green), neural/glial antigen NG-2 (H; green), α-SMA (H; red), Connexin-43 (I; green), Tuj-1 (I; magenta); growth cone (green; J), and NeuN (magenta, J). Bar charts represent the IMV (fluorescence) for corresponding protein as per randomly selected field area at the injury epicenter (3 fields/slide, n = 3/group). Data are mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 vs WT-injury group.

Discussion

In the present study, we establish that TRPV4, a nonselective cation channel expressed by astrocytes and neurons (Ryskamp et al., 2014; Shibasaki et al., 2014), is a crucial determinant of endothelial disruption and the pathology after SCI. We show that SCI induces the expression of TRPV4 at earlier time points; however, the free [Ca2+]i level follows a biphasic increase. However, there may be various mechanisms by which SCI produces these effects. (1) The increase in TRPV4 expression may be induced by mechanical stimulation as a result of cell swelling, as TRPV4 is also a volume-sensitive mechanosensor (Nilius et al., 2001; Shi et al., 2013). (2) Most Ca2+-permeable channels show some feedback regulation by Ca2+ to prevent deleteriously significant increases in [Ca2+]i or to shape the time course of channel activity, and experimental evidence shows that TRPV4 is tightly regulated by [Ca2+]i (Strotmann et al., 2003). (3) TRPV4 is highly sensitive to changes in extracellular osmolarity, as reductions in the extracellular osmolarity result in increases in [Ca2+]i and membrane currents and osmolarities >300 mosmol/L decrease both [Ca2+]i and currents (Strotmann et al., 2000). (4) Ca2+ is involved in both the decaying phase and the activation phase of TRPV4 (Plant and Strotmann, 2006). Traumatic SCI can result in contusion, compression, and stretch injury. Both compression and contusion models simulate the biomechanics and neuropathology of human injury (Sharif-Alhoseini et al., 2017). In the current study, we used clinically relevant SCI via controlled mechanical compression (Yu et al., 2013; Ropper et al., 2015). The advantage of the current method is that delivered force is precisely known at the time of application compared with other compression models. One limitation of the current methods is the variability in stabilizing and supporting the spinal column using Allis clamps at T8 and T12 spinous processes. Ideally, the spinal cord should be parallel to the impounder during compression to avoid inconsistent parenchymal injury. Second, the compression with this method is only applied to dorsally to the spinal cord.

We also found that the expression of TRPV4 at the injury epicenter, but not [Ca2+]i, progressively increases with the severity of the injury. TRPV4 does not have an apparent Ca2+-binding site, such as an EF-hand, suggesting that Ca2+ does not bind directly to the channel. Cells expressing TRPV4 often display spontaneous channel activity, resulting in an elevated basal [Ca2+]i (Plant and Strotmann, 2006), but there are also reports showing no changes in [Ca2+]i in cells expressing TRPV4 (Strotmann et al., 2000). Nonetheless, in the present study, [Ca2+]i was increased after injury, regardless of the severity. Thus, our findings suggest that TRPV4 expression might be useful for predicting the severity of the CNS injury. We also observed progressive damage to the vascular endothelium and neurons (via RECA and NF immunoreactivity, respectively). Interestingly, RECA immunoreactivity was low at DPI-1 when the expression of TRPV4 was high and matched the initial progressive damage to the endothelium, whereas axons showed progressive damage after SCI. These observations suggest that TRPV4 is increased and might promote the EC detachment following SCI.

Previous reports suggest that the expression of the TJ proteins occludin and ZO-1 is downregulated after moderate compression injury (Kumar et al., 2018c), suggesting a disruption of the BSCB. In the present study, the expression of occludin decreased while that of inflammatory cytokines (IL-6 and others) increased, suggesting the direct involvement of TRPV4 in BSCB disruption and inflammation. The upregulation of TRPV4 after SCI was also associated with a decrease in Pacsin-3, a synaptic vesicular membrane trafficking protein that inhibits TRPV4 basal expression (D'hoedt et al., 2008), suggesting that the upregulation of TRPV4 has functional relevance. TRPV4 is expressed by vascular ECs in rodents (Watanabe et al., 2002; Zhang et al., 2009) and humans (Sullivan et al., 2012) and has also been identified in vascular smooth muscle cells (Senadheera et al., 2013) and in the astrocytic endfoot processes that wrap around blood vessels in the CNS (Benfenati et al., 2011).TRPV4 activation within the astrocytic endfeet contributes to neurovascular coupling via Ca2+ entry (Dunn et al., 2011).

Adhesion, tight, and gap junctions connect the ECs lining the vessel walls (Cerutti and Ridley, 2017). In the current study, pharmacological activation of TRPV4 decreased the expression of TJ markers (ZO-1, ZO-2, and claudin-1) and remodel adhesion junctions (via afadin and endothelial actin-binding protein CD2-associated protein). In vivo application of a TRPV4 agonist led to EC damage and basal lamina deposition, which could diminish angiogenesis and promote cystic cavity formation, and enhanced EC damage after SCI. After SCI in rodents and humans, scarring occurs at the lesion site, comprising glial cells and laminin and fibronectin (Ruschel et al., 2015), which impedes axonal regeneration (Silver and Miller, 2004; Leal-Filho, 2011). However, the glial scars in animals receiving injections of the TRPV4 agonist were predominantly composed of microglia, which supports the notion that ion channels are also involved in regulating microglial functions (Färber and Kettenmann, 2005).

The activation of microglia after CNS injury is linked with neuroinflammation and impairments of the BBB/BSCB. TRPV4 activation also stimulates neuropeptide release from afferent nerves and induces neurogenic inflammation (Vergnolle et al., 2010). Whereas TRPV4 is activated by changes in osmolarity, which leads to increased IL-1β and IL-6 in intervertebral discs and is suggestive of increased release of proinflammatory cytokines (Walter et al., 2016), we found that inhibition of TRPV4 (via RN-1734) attenuated inflammation, reduced free [Ca2+]i levels, and preserved the BSCB.

The functional properties of TJ components and the selective leakiness of the CNS TJs are well studied (Bazzoni and Dejana, 2004; Abbott et al., 2010). Damage to these proteins leads to BSCB instability and increased permeability, which promotes inflammatory infiltration in the CNS, the major determinant of secondary injury (Noble and Wrathall, 1989). The temporal and spatial increase of secondary damage after SCI has been attributed to the augmented expression of sulfonylurea receptor 1-regulated NCCa-ATP channels, such as Trpm4, by ECs (Gerzanich et al., 2009). In the present study, we found that ECs in animals with TRPV4 deficiency were protected from damage after SCI, suggesting that the activation of TRPV4 after injury might directly affect EC survival. A possible explanation is that TRPV4-mediated entry of Ca2+ into ECs regulates the production of nitric oxide and responses to inflammatory signals by changing the barrier properties (Tiruppathi et al., 2002). For example, knockdown of TRPV4 in adipocytes is inhibitory for an array of cytokines and chemokines in adipose tissues, suggesting that TRPV4 positively controls proinflammatory genes (Ye et al., 2012). Consistent with this, we found that TRPV4 KO mice had reduced inflammation and synthesis of chemokines and cytokines after SCI as well as enhanced EC integrity, reduced permeability, and reduced apoptosis. After SCI, the expression of ANG-1 was reduced. ANG-1 lowers vascular leakage/inflammation and expedites angiogenesis (Lee et al., 2009), by strengthening related endothelial molecules and regulating interendothelial adhesion. We found that the reduction in ANG-1 was attenuated in TRPV4 KO mice. Additionally, TRPV4 KO mice maintained the levels of several neurotrophins (BDNF and NT-3), which are linked with EC survival (Donovan et al., 2000), as well as levels of vWF, NG-2, and SMA. Notably, the detachment of ECs, which persists throughout the acute injury phase (Koyanagi et al., 1993), is an indirect contributor to neuronal and glial cell death (Casella et al., 2002).

Inflammation is well established in the pathology of SCI, and contributes to fibrosis scarring in part via TGF-β signaling. The increase in TGF-β/CD-68 expression after SCI was attenuated in TRPV4 KO mice. We also observed an SCI-induced increase in astrocytes, microglia, extracellular matrix proteins, and gap junctions. The persistence of these glial cells indicates a proinflammatory environment, linked with increased neurotoxicity and impaired wound healing. However, these increases were attenuated in TRPV4 KO mice, suggesting that the increase in TRPV4 observed in WT animals enhanced the scarring process. Indeed, SCI-induced endothelial damage was augmented, and glial scar formation was higher in animals injected with the TRPV4 agonist. Scarring leads to the formation of a sharp lesion border, which impairs neuronal regeneration (Bundesen et al., 2003), by forming a physical barrier preventing regenerating axons from extending across the injury site. A reduction of one or many specific cellular components of the scar can influence the scarring (Meletis et al., 2008; Barnabé-Heider et al., 2010; Göritz et al., 2011; Zhu et al., 2015). Although TRPV4 also mediates neurotrophic factor-induced neuritogenesis in developing peripheral nerves (Jang et al., 2012), activation of this channel in the CNS via intracerebroventricular injection of the agonist induces neuronal apoptosis (Jie et al., 2015, 2016). Accordingly, we observed reduced immunoreactivity for NF, a cytoskeleton protein prominently expressed in larger axons, in animals injected with GSK, suggesting that TRPV4 activation affects large axons after SCI.

Finally, the sparing or regeneration of the vasculature and endothelial functions after injury correlates with improved functional outcomes (Kaneko et al., 2006). The above-described reduction in glial scarring and inflammation and enhanced expression of neuronal markers in KO mice suggest that TRPV4 contributes to the pathology of, and impaired recovery from, SCI. Accordingly, we observed reduced hyperalgesia toward heat and faster locomotor recovery in TRPV4 KO mice after SCI. The observed functional recovery might be attributed to reduced reactive gliosis. However, TRPV4 deficiency does not affect escape latency in response to heat in the absence of hyperalgesia (Liedtke and Friedman, 2003; Suzuki et al., 2003; Todaka et al., 2004), although an increase in tail withdrawal latency to moderately hot temperatures was reported (Lee et al., 2005). Other studies suggest that the channel contributes to the sensation of noxious mechanical stimuli (Liedtke and Friedman, 2003; Suzuki et al., 2003). Together, these data suggest that TRPV4 contributes to nociception only in the setting of inflammation or nerve injury (Alessandri-Haber et al., 2008).

In conclusion, TRPV4 expression is rapidly induced following SCI and in accordance with the severity of the injury. Its expression is linked to endothelial damage, inflammation, and apoptosis, thereby increasing secondary injury. Importantly, inhibition/knockdown of TRPV4 prevented these effects. Thus, the manipulation of TRPV4 signaling might lead to new therapeutic strategies or combinatorial therapies to protect ECs and enhance repair after SCI.

Footnotes

  • This work was supported by National Research Foundation of Korea Grants NRF-2015H1D3A1066543, NRF-2017R1C1B2011772, NRF 2017R1C1B1011397 and NRF-2019R1A5A2026045, Korea Healthcare Technology Research&Development Project, and Ministry for Health & Welfare Affairs, Republic of Korea HR16C0002 and HI18C0183.

  • The authors declare no competing financial interests.

  • Correspondence should be addressed to In-Bo Han at hanib{at}cha.ac.kr

References

  1. ↵
    1. Abbott NJ,
    2. Patabendige AA,
    3. Dolman DE,
    4. Yusof SR,
    5. Begley DJ
    (2010) Structure and function of the blood–brain barrier. Neurobiol Dis 37:13–25. doi:10.1016/j.nbd.2009.07.030 pmid:19664713
    OpenUrlCrossRefPubMed
  2. ↵
    1. Alessandri-Haber N,
    2. Dina OA,
    3. Yeh JJ,
    4. Parada CA,
    5. Reichling DB,
    6. Levine JD
    (2004) Transient receptor potential vanilloid 4 is essential in chemotherapy-induced neuropathic pain in the rat. J Neurosci 24:4444–4452. doi:10.1523/JNEUROSCI.0242-04.2004 pmid:15128858
    OpenUrlAbstract/FREE Full Text
  3. ↵
    1. Alessandri-Haber N,
    2. Dina OA,
    3. Joseph EK,
    4. Reichling D,
    5. Levine JD
    (2006) A transient receptor potential vanilloid 4-dependent mechanism of hyperalgesia is engaged by concerted action of inflammatory mediators. J Neurosci 26:3864–3874. doi:10.1523/JNEUROSCI.5385-05.2006 pmid:16597741
    OpenUrlAbstract/FREE Full Text
  4. ↵
    1. Alessandri-Haber N,
    2. Dina OA,
    3. Joseph EK,
    4. Reichling DB,
    5. Levine JD
    (2008) Interaction of transient receptor potential vanilloid 4, integrin, and SRC tyrosine kinase in mechanical hyperalgesia. J Neurosci 28:1046–1057. doi:10.1523/JNEUROSCI.4497-07.2008 pmid:18234883
    OpenUrlAbstract/FREE Full Text
  5. ↵
    1. Barnabé-Heider F,
    2. Göritz C,
    3. Sabelström H,
    4. Takebayashi H,
    5. Pfrieger FW,
    6. Meletis K,
    7. Frisén J
    (2010) Origin of new glial cells in intact and injured adult spinal cord. Cell Stem Cell 7:470–482. doi:10.1016/j.stem.2010.07.014 pmid:20887953
    OpenUrlCrossRefPubMed
  6. ↵
    1. Basso DM,
    2. Fisher LC,
    3. Anderson AJ,
    4. Jakeman LB,
    5. McTigue DM,
    6. Popovich PG
    (2006) Basso mouse scale for locomotion detects differences in recovery after spinal cord injury in five common mouse strains. J Neurotrauma 23:635–659. doi:10.1089/neu.2006.23.635 pmid:16689667
    OpenUrlCrossRefPubMed
  7. ↵
    1. Bazzoni G,
    2. Dejana E
    (2004) Endothelial cell-to-cell junctions: molecular organization and role in vascular homeostasis. Physiol Rev 84:869–901. doi:10.1152/physrev.00035.2003 pmid:15269339
    OpenUrlCrossRefPubMed
  8. ↵
    1. Benfenati V,
    2. Caprini M,
    3. Dovizio M,
    4. Mylonakou MN,
    5. Ferroni S,
    6. Ottersen OP,
    7. Amiry-Moghaddam M
    (2011) An aquaporin-4/transient receptor potential vanilloid 4 (AQP4/TRPV4) complex is essential for cell-volume control in astrocytes. Proc Natl Acad Sci U S A 108:2563–2568. doi:10.1073/pnas.1012867108 pmid:21262839
    OpenUrlAbstract/FREE Full Text
  9. ↵
    1. Benton RL,
    2. Maddie MA,
    3. Worth CA,
    4. Mahoney ET,
    5. Hagg T,
    6. Whittermore SR
    (2008) Transcriptomic screening of microvascular endothelial cells implicates novel molecular regulators of vascular dysfunction after spinal cord injury. J Cereb Blood Flow Metab 28:1771–1785. doi:10.1038/jcbfm.2008.76 pmid:18612314
    OpenUrlCrossRefPubMed
  10. ↵
    1. Bundesen LQ,
    2. Scheel TA,
    3. Bregman BS,
    4. Kromer LF
    (2003) Ephrin-B2 and EphB2 regulation of astrocyte-meningeal fibroblast interactions in response to spinal cord lesions in adult rats. J Neurosci 23:7789–7800. doi:10.1523/JNEUROSCI.23-21-07789.2003 pmid:12944508
    OpenUrlAbstract/FREE Full Text
  11. ↵
    1. Casella GT,
    2. Marcillo A,
    3. Bunge MB,
    4. Wood PM
    (2002) New vascular tissue rapidly replaces neural parenchyma and vessels destroyed by a contusion injury to the rat spinal cord. Exp Neurol 173:63–76. doi:10.1006/exnr.2001.7827 pmid:11771939
    OpenUrlCrossRefPubMed
  12. ↵
    1. Casella GT,
    2. Bunge MB,
    3. Wood PM
    (2006) Endothelial cell loss is not a major cause of neuronal and glial cell death following contusion injury of the spinal cord. Exp Neurol 202:8–20. doi:10.1016/j.expneurol.2006.05.028 pmid:16872600
    OpenUrlCrossRefPubMed
  13. ↵
    1. Cerutti C,
    2. Ridley AJ
    (2017) Endothelial cell–cell adhesion and signaling. Exp Cell Res 358:31–38. doi:10.1016/j.yexcr.2017.06.003 pmid:28602626
    OpenUrlCrossRefPubMed
  14. ↵
    1. D'hoedt D,
    2. Owsianik G,
    3. Prenen J,
    4. Cuajungco MP,
    5. Grimm C,
    6. Heller S,
    7. Voets T,
    8. Nilius B
    (2008) Stimulus-specific modulation of the cation channel TRPV4 by PACSIN 3. J Biol Chem 283:6272–6280. doi:10.1074/jbc.M706386200 pmid:18174177
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Donovan MJ,
    2. Lin MI,
    3. Wiegn P,
    4. Ringstedt T,
    5. Kraemer R,
    6. Hahn R,
    7. Wang S,
    8. Ibañez CF,
    9. Rafii S,
    10. Hempstead BL
    (2000) Brain derived neurotrophic factor is an endothelial cell survival factor required for intramyocardial vessel stabilization. Development 127:4531–4540. pmid:11023857
    OpenUrlAbstract
  16. ↵
    1. Dunn KM,
    2. Bonev AD,
    3. Nelson MT
    (2011) Functional evidence of TRPV4-mediated Ca2+ signals in cortical astrocytes. FASEB J 25:1023–1024.
    OpenUrl
  17. ↵
    1. Färber K,
    2. Kettenmann H
    (2005) Physiology of microglial cells. Brain Res Brain Res Rev 48:133–143. doi:10.1016/j.brainresrev.2004.12.003 pmid:15850652
    OpenUrlCrossRefPubMed
  18. ↵
    1. Fassbender JM,
    2. Whittemore SR,
    3. Hagg T
    (2011) Targeting microvasculature for neuroprotection after SCI. Neurotherapeutics 8:240–251. doi:10.1007/s13311-011-0029-1 pmid:21360237
    OpenUrlCrossRefPubMed
  19. ↵
    1. Gerzanich V,
    2. Woo SK,
    3. Vennekens R,
    4. Tsymbalyuk O,
    5. Ivanova S,
    6. Ivanov A,
    7. Geng Z,
    8. Chen Z,
    9. Nilius B,
    10. Flockerzi V,
    11. Freichel M,
    12. Simard JM
    (2009) De novo expression of Trpm4 initiates secondary hemorrhage in spinal cord injury. Nat Med 15:185–191. doi:10.1038/nm.1899 pmid:19169264
    OpenUrlCrossRefPubMed
  20. ↵
    1. Göritz C,
    2. Dias DO,
    3. Tomilin N,
    4. Barbacid M,
    5. Shupliakov O,
    6. Frisén J
    (2011) A pericyte origin of spinal cord scar tissue. Science 333:238–242. doi:10.1126/science.1203165 pmid:21737741
    OpenUrlAbstract/FREE Full Text
  21. ↵
    1. Grant AD,
    2. Cottrell GS,
    3. Amadesi S,
    4. Trevisani M,
    5. Nicoletti P,
    6. Materazzi S,
    7. Altier C,
    8. Cenac N,
    9. Zamponi GW,
    10. Bautista-Cruz F,
    11. Lopez CB,
    12. Joseph EK,
    13. Levine JD,
    14. Liedtke W,
    15. Vanner S,
    16. Vergnolle N,
    17. Geppetti P,
    18. Bunnett NW
    (2007) Protease-activated receptor 2 sensitizes the transient receptor potential vanilloid 4 ion channel to cause mechanical hyperalgesia in mice. J Physiol 578:715–733. doi:10.1113/jphysiol.2006.121111 pmid:17124270
    OpenUrlCrossRefPubMed
  22. ↵
    1. Guo S,
    2. Kim WJ,
    3. Lok J,
    4. Lee SR,
    5. Besancon E,
    6. Luo BH,
    7. Stins MF,
    8. Wang X,
    9. Dedhar S,
    10. Lo EH
    (2008) Neuroprotection via matrix-trophic coupling between cerebral endothelial cells and neurons. Proc Natl Acad Sci U S A 105:7582–7587. doi:10.1073/pnas.0801105105 pmid:18495934
    OpenUrlAbstract/FREE Full Text
  23. ↵
    1. Jang Y,
    2. Jung J,
    3. Kim H,
    4. Oh J,
    5. Jeon JH,
    6. Jung S,
    7. Kim KT,
    8. Cho H,
    9. Yang DJ,
    10. Kim SM,
    11. Kim IB,
    12. Song MR,
    13. Oh U
    (2012) Axonal neuropathy-associated TRPV4 regulates neurotrophic factor-derived axonal growth. J Biol Chem 287:6014–6024. doi:10.1074/jbc.M111.316315 pmid:22187434
    OpenUrlAbstract/FREE Full Text
  24. ↵
    1. Jie P,
    2. Hong Z,
    3. Tian Y,
    4. Li Y,
    5. Lin L,
    6. Zhou L,
    7. Du Y,
    8. Chen L,
    9. Chen L
    (2015) Activation of transient receptor potential vanilloid 4 induces apoptosis in hippocampus through downregulating PI3K/Akt and upregulating p38 MAPK signaling pathways. Cell Death Dis 6:e1775. doi:10.1038/cddis.2015.146 pmid:26043075
    OpenUrlCrossRefPubMed
  25. ↵
    1. Jie P,
    2. Lu Z,
    3. Hong Z,
    4. Li L,
    5. Zhou L,
    6. Li Y,
    7. Zhou R,
    8. Zhou Y,
    9. Du Y,
    10. Chen L,
    11. Chen L
    (2016) Activation of transient receptor potential vanilloid 4 is involved in neuronal injury in middle cerebral artery occlusion in mice. Mol Neurobiol 53:8–17. doi:10.1007/s12035-014-8992-2 pmid:25399955
    OpenUrlCrossRefPubMed
  26. ↵
    1. Kaneko S,
    2. Iwanami A,
    3. Nakamura M,
    4. Kishino A,
    5. Kikuchi K,
    6. Shibata S,
    7. Okano HJ,
    8. Ikegami T,
    9. Moriya A,
    10. Konishi O,
    11. Nakayama C,
    12. Kumagai K,
    13. Kimura T,
    14. Sato Y,
    15. Goshima Y,
    16. Taniguchi M,
    17. Ito M,
    18. He Z,
    19. Toyama Y,
    20. Okano H
    (2006) A selective Sema3A inhibitor enhances regenerative responses and functional recovery of the injured spinal cord. Nat Med 12:1380–1389. doi:10.1038/nm1505 pmid:17099709
    OpenUrlCrossRefPubMed
  27. ↵
    1. Kim HJ,
    2. Lim CS,
    3. Lee HW,
    4. Lee HS,
    5. Um YJ,
    6. Kumar H,
    7. Han I,
    8. Kim HM
    (2017) A ratiometric two-photon probe for Ca2+ in live tissues and its application to spinal cord injury model. Biomaterials 141:251–259. doi:10.1016/j.biomaterials.2017.07.006 pmid:28697466
    OpenUrlCrossRefPubMed
  28. ↵
    1. Koyanagi I,
    2. Tator CH,
    3. Lea PJ
    (1993) Three-dimensional analysis of the vascular system in the rat spinal cord with scanning electron microscopy of vascular corrosion casts: 2. Acute spinal cord injury. Neurosurgery 33:285–292. doi:10.1097/00006123-199308000-00016 pmid:8367052
    OpenUrlCrossRefPubMed
  29. ↵
    1. Kumar H,
    2. Lee SH,
    3. Kim KT,
    4. Zeng X,
    5. Han I
    (2018a) TRPV4: a sensor for homeostasis and pathological events in the CNS. Mol Neurobiol 55:8695–8708. doi:10.1007/s12035-018-0998-8 pmid:29582401
    OpenUrlCrossRefPubMed
  30. ↵
    1. Kumar H,
    2. Jo MJ,
    3. Choi H,
    4. Muttigi MS,
    5. Shon S,
    6. Kim BJ,
    7. Lee SH,
    8. Han IB
    (2018b) Matrix metalloproteinase-8 inhibition prevents disruption of blood–spinal cord barrier and attenuates inflammation in rat model of spinal cord injury. Mol Neurobiol 55:2577–2590. doi:10.1007/s12035-017-0509-3 pmid:28421532
    OpenUrlCrossRefPubMed
  31. ↵
    1. Kumar H,
    2. Choi H,
    3. Jo MJ,
    4. Joshi HP,
    5. Muttigi M,
    6. Bonanomi D,
    7. Kim SB,
    8. Ban E,
    9. Kim A,
    10. Lee SH,
    11. Kim KT,
    12. Sohn S,
    13. Zeng X,
    14. Han I
    (2018c) Neutrophil elastase inhibition effectively rescued angiopoietin-1 decrease and inhibits glial scar after spinal cord injury. Acta Neuropathol Commun 6:73. doi:10.1186/s40478-018-0576-3 pmid:30086801
    OpenUrlCrossRefPubMed
  32. ↵
    1. Leal-Filho MB
    (2011) Spinal cord injury: from inflammation to glial scar. Surg Neurol Int 2:112. doi:10.4103/2152-7806.83732 pmid:21886885
    OpenUrlCrossRefPubMed
  33. ↵
    1. Lee HS,
    2. Han J,
    3. Bai HJ,
    4. Kim KW
    (2009) Brain angiogenesis in developmental and pathological processes: regulation, molecular and cellular communication at the neurovascular interface. FEBS J 276:4622–4635. doi:10.1111/j.1742-4658.2009.07174.x pmid:19664072
    OpenUrlCrossRefPubMed
  34. ↵
    1. Lee H,
    2. Iida T,
    3. Mizuno A,
    4. Suzuki M,
    5. Caterina MJ
    (2005) Altered thermal selection behavior in mice lacking transient receptor potential vanilloid 4. J Neurosci 25:1304–1310. doi:10.1523/JNEUROSCI.4745.04.2005 pmid:15689568
    OpenUrlAbstract/FREE Full Text
  35. ↵
    1. Liedtke W,
    2. Friedman JM
    (2003) Abnormal osmotic regulation in trpv4−/− mice. Proc Natl Acad Sci U S A 100:13698–13703. doi:10.1073/pnas.1735416100 pmid:14581612
    OpenUrlAbstract/FREE Full Text
  36. ↵
    1. Liedtke W,
    2. Choe Y,
    3. Martí-Renom MA,
    4. Bell AM,
    5. Denis CS,
    6. Sali A,
    7. Hudspeth AJ,
    8. Friedman JM,
    9. Heller S
    (2000) Vanilloid receptor-related osmotically activated channel (VR-OAC), a candidate vertebrate osmoreceptor. Cell 103:525–535. doi:10.1016/S0092-8674(00)00143-4 pmid:11081638
    OpenUrlCrossRefPubMed
  37. ↵
    1. Ling X,
    2. Liu D
    (2007) Temporal and spatial profiles of cell loss after spinal cord injury: reduction by a metalloporphyrin. J Neurosci Res 85:2175–2185. doi:10.1002/jnr.21362 pmid:17551979
    OpenUrlCrossRefPubMed
  38. ↵
    1. Liu XZ,
    2. Xu XM,
    3. Hu R,
    4. Du C,
    5. Zhang SX,
    6. McDonald JW,
    7. Dong HX,
    8. Wu YJ,
    9. Fan GS,
    10. Jacquin MF,
    11. Hsu CY,
    12. Choi DW
    (1997) Neuronal and glial apoptosis after traumatic spinal cord injury. J Neurosci 17:5395–5406. doi:10.1523/JNEUROSCI.17-14-05395.1997 pmid:9204923
    OpenUrlAbstract/FREE Full Text
  39. ↵
    1. Meletis K,
    2. Barnabé-Heider F,
    3. Carlén M,
    4. Evergren E,
    5. Tomilin N,
    6. Shupliakov O,
    7. Frisén J
    (2008) Spinal cord injury reveals multilineage differentiation of ependymal cells. PLoS Biol 6:e182. doi:10.1371/journal.pbio.0060182 pmid:18651793
    OpenUrlCrossRefPubMed
  40. ↵
    1. Montell C,
    2. Birnbaumer L,
    3. Flockerzi V
    (2002) The TRP channels, a remarkably functional family. Cell 108:595–598. doi:10.1016/S0092-8674(02)00670-0 pmid:11893331
    OpenUrlCrossRefPubMed
  41. ↵
    1. Narita K,
    2. Sasamoto S,
    3. Koizumi S,
    4. Okazaki S,
    5. Nakamura H,
    6. Inoue T,
    7. Takeda S
    (2015) TRPV4 regulates the integrity of the blood-cerebrospinal fluid barrier and modulates transepithelial protein transport. FASEB J 29:2247–2259. doi:10.1096/fj.14-261396 pmid:25681460
    OpenUrlCrossRefPubMed
  42. ↵
    National Research Council (2011) Guide for the care and use of laboratory animals. Washington, DC: National Research Council.
  43. ↵
    1. Nilius B,
    2. Droogmans G
    (2001) Ion channels and their functional role in vascular endothelium. Physiol Rev 81:1415–1459. doi:10.1152/physrev.2001.81.4.1415 pmid:11581493
    OpenUrlCrossRefPubMed
  44. ↵
    1. Nilius B,
    2. Prenen J,
    3. Wissenbach U,
    4. Bödding M,
    5. Droogmans G
    (2001) Differential activation of the volume-sensitive cation channel TRP12 (OTRPC4) and volume-regulated anion currents in HEK-293 cells. Pflugers Archiv 443:227–233. doi:10.1007/s004240100676 pmid:11713648
    OpenUrlCrossRefPubMed
  45. ↵
    1. Noble LJ,
    2. Wrathall JR
    (1989) Distribution and time course of protein extravasation in the rat spinal cord after contusive injury. Brain Res 482:57–66. doi:10.1016/0006-8993(89)90542-8 pmid:2706482
    OpenUrlCrossRefPubMed
  46. ↵
    1. Plant TD,
    2. Strotmann R
    (2006) TRPV4: a multifunctional nonselective cation channel with complex regulation. In: TRP ion channel function in sensory transduction and cellular signaling cascades, pp 139–154. Boca Raton, FL: CRC.
  47. ↵
    1. Ramsey IS,
    2. Delling M,
    3. Clapham DE
    (2006) An introduction to TRP channels. Annu Rev Physiol 68:619–647. doi:10.1146/annurev.physiol.68.040204.100431 pmid:16460286
    OpenUrlCrossRefPubMed
  48. ↵
    1. Ropper AE,
    2. Zeng X,
    3. Anderson JE,
    4. Yu D,
    5. Han I,
    6. Haragopal H,
    7. Teng YD
    (2015) An efficient device to experimentally model compression injury of mammalian spinal cord. Exp Neurol 271:515–523. doi:10.1016/j.expneurol.2015.07.012 pmid:26210871
    OpenUrlCrossRefPubMed
  49. ↵
    1. Ruschel J,
    2. Hellal F,
    3. Flynn KC,
    4. Dupraz S,
    5. Elliott DA,
    6. Tedeschi A,
    7. Bates M,
    8. Sliwinski C,
    9. Brook G,
    10. Dobrindt K,
    11. Peitz M,
    12. Brüstle O,
    13. Norenberg MD,
    14. Blesch A,
    15. Weidner N,
    16. Bunge MB,
    17. Bixby JL,
    18. Bradke F
    (2015) Axonal regeneration. systemic administration of epothilone B promotes axon regeneration after spinal cord injury. Science 348:347–352. doi:10.1126/science.aaa2958 pmid:25765066
    OpenUrlAbstract/FREE Full Text
  50. ↵
    1. Ryskamp DA,
    2. Jo AO,
    3. Frye AM,
    4. Vazquez-Chona F,
    5. MacAulay N,
    6. Thoreson WB,
    7. Krizaj D
    (2014) Swelling and eicosanoid metabolites differentially gate TRPV4 channels in retinal neurons and glia. J Neurosci 34:15689–15700. doi:10.1523/JNEUROSCI.2540-14.2014 pmid:25411497
    OpenUrlAbstract/FREE Full Text
  51. ↵
    1. Senadheera S,
    2. Bertrand PP,
    3. Grayson TH,
    4. Leader L,
    5. Murphy TV,
    6. Sandow SL
    (2013) Pregnancy-induced remodelling and enhanced endothelium-derived hyperpolarization-type vasodilator activity in rat uterine radial artery: transient receptor potential vanilloid type 4 channels, caveolae and myoendothelial gap junctions. J Anat 223:677–686. doi:10.1111/joa.12127 pmid:24128141
    OpenUrlCrossRefPubMed
  52. ↵
    1. Sharif-Alhoseini M,
    2. Khormali M,
    3. Rezaei M,
    4. Safdarian M,
    5. Hajighadery A,
    6. Khalatbari MM,
    7. Meknatkhah S,
    8. Rezvan M,
    9. Chalangari M,
    10. Derakhshan P,
    11. Rahimi-Movaghar V
    (2017) Animal models of spinal cord injury: a systematic review. Spinal Cord 55:714–721. doi:10.1038/sc.2016.187 pmid:28117332
    OpenUrlCrossRefPubMed
  53. ↵
    1. Shi M,
    2. Du F,
    3. Liu Y,
    4. Li L,
    5. Cai J,
    6. Zhang GF,
    7. Xu XF,
    8. Lin T,
    9. Cheng HR,
    10. Liu XD,
    11. Xiong LZ,
    12. Zhao G
    (2013) Glial cell-expressed mechanosensitive channel TRPV4 mediates infrasound-induced neuronal impairment. Acta Neuropathol 126:725–739. doi:10.1007/s00401-013-1166-x pmid:24002225
    OpenUrlCrossRefPubMed
  54. ↵
    1. Shibasaki K,
    2. Ikenaka K,
    3. Tamalu F,
    4. Tominaga M,
    5. Ishizaki Y
    (2014) A novel subtype of astrocytes expressing TRPV4 (transient receptor potential vanilloid 4) regulates neuronal excitability via release of gliotransmitters. J Biol Chem 289:14470–14480. doi:10.1074/jbc.M114.557132 pmid:24737318
    OpenUrlAbstract/FREE Full Text
  55. ↵
    1. Silver J,
    2. Miller JH
    (2004) Regeneration beyond the glial scar. Nat Rev Neurosci 5:146–156. doi:10.1038/nrn1326 pmid:14735117
    OpenUrlCrossRefPubMed
  56. ↵
    1. Strotmann R,
    2. Harteneck C,
    3. Nunnenmacher K,
    4. Schultz G,
    5. Plant TD
    (2000) OTRPC4, a nonselective cation channel that confers sensitivity to extracellular osmolarity. Nat Cell Biol 2:695–702. doi:10.1038/35036318 pmid:11025659
    OpenUrlCrossRefPubMed
  57. ↵
    1. Strotmann R,
    2. Schultz G,
    3. Plant TD
    (2003) Ca2+-dependent potentiation of the nonselective cation channel TRPV4 is mediated by a C-terminal calmodulin binding site. J Biol Chem 278:26541–26549. doi:10.1074/jbc.M302590200 pmid:12724311
    OpenUrlAbstract/FREE Full Text
  58. ↵
    1. Sullivan MN,
    2. Francis M,
    3. Pitts NL,
    4. Taylor MS,
    5. Earley S
    (2012) Optical recording reveals novel properties of GSK1016790A-induced vanilloid transient receptor potential channel TRPV4 activity in primary human endothelial cells. Mol Pharmacol 82:464–472. doi:10.1124/mol.112.078584 pmid:22689561
    OpenUrlAbstract/FREE Full Text
  59. ↵
    1. Suzuki M,
    2. Mizuno A,
    3. Kodaira K,
    4. Imai M
    (2003) Impaired pressure sensation in mice lacking TRPV4. J Biol Chem 278:22664–22668. doi:10.1074/jbc.M302561200 pmid:12692122
    OpenUrlAbstract/FREE Full Text
  60. ↵
    1. Tiruppathi C,
    2. Freichel M,
    3. Vogel SM,
    4. Paria BC,
    5. Mehta D,
    6. Flockerzi V,
    7. Malik AB
    (2002) Impairment of store-operated Ca2+ entry in TRPC4−/− mice interferes with increase in lung microvascular permeability. Circ Res 91:70–76. doi:10.1161/01.RES.0000023391.40106.A8 pmid:12114324
    OpenUrlAbstract/FREE Full Text
  61. ↵
    1. Todaka H,
    2. Taniguchi J,
    3. Satoh J,
    4. Mizuno A,
    5. Suzuki M
    (2004) Warm temperature-sensitive transient receptor potential vanilloid 4 (TRPV4) plays an essential role in thermal hyperalgesia. J Biol Chem 279:35133–35138. doi:10.1074/jbc.M406260200 pmid:15187078
    OpenUrlAbstract/FREE Full Text
  62. ↵
    1. Vergnolle N,
    2. Cenac N,
    3. Altier C,
    4. Cellars L,
    5. Chapman K,
    6. Zamponi GW,
    7. Materazzi S,
    8. Nassini R,
    9. Liedtke W,
    10. Cattaruzza F,
    11. Grady EF,
    12. Geppetti P,
    13. Bunnett NW
    (2010) A role for transient receptor potential vanilloid 4 in tonicity-induced neurogenic inflammation. Br J Pharmacol 159:1161–1173. doi:10.1111/j.1476-5381.2009.00590.x pmid:20136846
    OpenUrlCrossRefPubMed
  63. ↵
    1. Walter BA,
    2. Purmessur D,
    3. Moon A,
    4. Occhiogrosso J,
    5. Laudier DM,
    6. Hecht AC,
    7. Iatridis JC
    (2016) Reduced tissue osmolarity increases TRPV4 expression and pro-inflammatory cytokines in intervertebral disc cells. Eur Cell Mater 32:123–136. doi:10.22203/eCM.v032a08 pmid:27434269
    OpenUrlCrossRefPubMed
  64. ↵
    1. Watanabe H,
    2. Vriens J,
    3. Suh SH,
    4. Benham CD,
    5. Droogmans G,
    6. Nilius B
    (2002) Heat-evoked activation of TRPV4 channels in a HEK293 cell expression system and in native mouse aorta endothelial cells. J Biol Chem 277:47044–47051. doi:10.1074/jbc.M208277200 pmid:12354759
    OpenUrlAbstract/FREE Full Text
  65. ↵
    1. Whetstone WD,
    2. Hsu JY,
    3. Eisenberg M,
    4. Werb Z,
    5. Noble-Haeusslein LJ
    (2003) Blood–spinal cord barrier after spinal cord injury: relation to revascularization and wound healing. J Neurosci Res 74:227–239. doi:10.1002/jnr.10759 pmid:14515352
    OpenUrlCrossRefPubMed
  66. ↵
    1. Willette RN,
    2. Bao W,
    3. Nerurkar S,
    4. Yue TL,
    5. Doe CP,
    6. Stankus G,
    7. Turner GH,
    8. Ju H,
    9. Thomas H,
    10. Fishman CE,
    11. Sulpizio A,
    12. Behm DJ,
    13. Hoffman S,
    14. Lin Z,
    15. Lozinskaya I,
    16. Casillas LN,
    17. Lin M,
    18. Trout RE,
    19. Votta BJ,
    20. Thorneloe K, et al
    . (2008) Systemic activation of the transient receptor potential vanilloid subtype 4 channel causes endothelial failure and circulatory collapse: part 2. J Pharmacol Exp Ther 326:443–452. doi:10.1124/jpet.107.134551 pmid:18499744
    OpenUrlAbstract/FREE Full Text
  67. ↵
    1. Ye L,
    2. Kleiner S,
    3. Wu J,
    4. Sah R,
    5. Gupta RK,
    6. Banks AS,
    7. Cohen P,
    8. Khandekar MJ,
    9. Boström P,
    10. Mepani RJ,
    11. Laznik D,
    12. Kamenecka TM,
    13. Song X,
    14. Liedtke W,
    15. Mootha VK,
    16. Puigserver P,
    17. Griffin PR,
    18. Clapham DE,
    19. Spiegelman BM
    (2012) TRPV4 is a regulator of adipose oxidative metabolism, inflammation, and energy homeostasis. Cell 151:96–110. doi:10.1016/j.cell.2012.08.034 pmid:23021218
    OpenUrlCrossRefPubMed
  68. ↵
    1. Yu D,
    2. Thakor DK,
    3. Han I,
    4. Ropper AE,
    5. Haragopal H,
    6. Sidman RL,
    7. Zafonte R,
    8. Schachter SC,
    9. Teng YD
    (2013) Alleviation of chronic pain following rat spinal cord compression injury with multimodal actions of huperzine A. Proc Natl Acad Sci U S A 110:E746–E755. doi:10.1073/pnas.1300083110 pmid:23386718
    OpenUrlAbstract/FREE Full Text
  69. ↵
    1. Zhang DX,
    2. Mendoza SA,
    3. Bubolz AH,
    4. Mizuno A,
    5. Ge ZD,
    6. Li R,
    7. Warltier DC,
    8. Suzuki M,
    9. Gutterman DD
    (2009) Transient receptor potential vanilloid type 4-deficient mice exhibit impaired endothelium-dependent relaxation induced by acetylcholine in vitro and in vivo. Hypertension 53:532–538. doi:10.1161/HYPERTENSIONAHA.108.127100 pmid:19188524
    OpenUrlCrossRefPubMed
  70. ↵
    1. Zhao P,
    2. Lieu T,
    3. Barlow N,
    4. Metcalf M,
    5. Veldhuis NA,
    6. Jensen DD,
    7. Kocan M,
    8. Sostegni S,
    9. Haerteis S,
    10. Baraznenok V,
    11. Henderson I,
    12. Lindström E,
    13. Guerrero-Alba R,
    14. Valdez-Morales EE,
    15. Liedtke W,
    16. McIntyre P,
    17. Vanner SJ,
    18. Korbmacher C,
    19. Bunnett NW
    (2014) Cathepsin S causes inflammatory pain via biased agonism of PAR2 and TRPV4. J Biol Chem 289:27215–27234. doi:10.1074/jbc.M114.599712 pmid:25118282
    OpenUrlAbstract/FREE Full Text
  71. ↵
    1. Zhu Y,
    2. Soderblom C,
    3. Trojanowsky M,
    4. Lee DH,
    5. Lee JK
    (2015) Fibronectin matrix assembly after spinal cord injury. J Neurotrauma 32:1158–1167. doi:10.1089/neu.2014.3703 pmid:25492623
    OpenUrlCrossRefPubMed
Back to top

In this issue

The Journal of Neuroscience: 40 (9)
Journal of Neuroscience
Vol. 40, Issue 9
26 Feb 2020
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Advertising (PDF)
  • Ed Board (PDF)
Email

Thank you for sharing this Journal of Neuroscience article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Elevated TRPV4 Levels Contribute to Endothelial Damage and Scarring in Experimental Spinal Cord Injury
(Your Name) has forwarded a page to you from Journal of Neuroscience
(Your Name) thought you would be interested in this article in Journal of Neuroscience.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
View Full Page PDF
Citation Tools
Elevated TRPV4 Levels Contribute to Endothelial Damage and Scarring in Experimental Spinal Cord Injury
Hemant Kumar, Chang Su Lim, Hyemin Choi, Hari Prasad Joshi, Kyoung-Tae Kim, Yong Ho Kim, Chul-Kyu Park, Hwan Myung Kim, In-Bo Han
Journal of Neuroscience 26 February 2020, 40 (9) 1943-1955; DOI: 10.1523/JNEUROSCI.2035-19.2020

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Respond to this article
Request Permissions
Share
Elevated TRPV4 Levels Contribute to Endothelial Damage and Scarring in Experimental Spinal Cord Injury
Hemant Kumar, Chang Su Lim, Hyemin Choi, Hari Prasad Joshi, Kyoung-Tae Kim, Yong Ho Kim, Chul-Kyu Park, Hwan Myung Kim, In-Bo Han
Journal of Neuroscience 26 February 2020, 40 (9) 1943-1955; DOI: 10.1523/JNEUROSCI.2035-19.2020
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Keywords

  • blood–spinal cord barrier
  • endothelial cell
  • scarring
  • spinal cord injury
  • TRPV4
  • two-photon microscopy

Responses to this article

Respond to this article

Jump to comment:

No eLetters have been published for this article.

Related Articles

Cited By...

More in this TOC Section

Research Articles

  • Vocal error monitoring in the primate auditory cortex
  • CaMKIIβ-mediated phosphorylation enhances protein stability of spastin to promote neurite outgrowth
  • Neural Distinction between Visual Word and Object Recognition: An fMRI Study Using Pictographs
Show more Research Articles

Neurobiology of Disease

  • Unveiling cortical criticality changes along the prodromal to the overt continuum of alpha-synucleinopathy
  • Atypical Retinal Ganglion Cell Function in a Mouse Model of Fragile X Syndrome
  • Basal forebrain-ventral tegmental area glutamatergic pathway promotes emergence from isoflurane anesthesia in mice
Show more Neurobiology of Disease
  • Home
  • Alerts
  • Follow SFN on BlueSky
  • Visit Society for Neuroscience on Facebook
  • Follow Society for Neuroscience on Twitter
  • Follow Society for Neuroscience on LinkedIn
  • Visit Society for Neuroscience on Youtube
  • Follow our RSS feeds

Content

  • Early Release
  • Current Issue
  • Issue Archive
  • Collections

Information

  • For Authors
  • For Advertisers
  • For the Media
  • For Subscribers

About

  • About the Journal
  • Editorial Board
  • Privacy Notice
  • Contact
  • Accessibility
(JNeurosci logo)
(SfN logo)

Copyright © 2025 by the Society for Neuroscience.
JNeurosci Online ISSN: 1529-2401

The ideas and opinions expressed in JNeurosci do not necessarily reflect those of SfN or the JNeurosci Editorial Board. Publication of an advertisement or other product mention in JNeurosci should not be construed as an endorsement of the manufacturer’s claims. SfN does not assume any responsibility for any injury and/or damage to persons or property arising from or related to any use of any material contained in JNeurosci.