51
|
Wang Q, Duan M, Liao J, Xie J, Zhou C. Are Osteoclasts Mechanosensitive Cells? J Biomed Nanotechnol 2021; 17:1917-1938. [PMID: 34706793 DOI: 10.1166/jbn.2021.3171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Skeleton metabolism is a process in which osteoclasts constantly remove old bone and osteoblasts form new osteoid and induce mineralization; disruption of this balance may cause diseases. Osteoclasts play a key role in bone metabolism, as osteoclastogenesis marks the beginning of each bone remodeling cycle. As the only cell capable of bone resorption, osteoclasts are derived from the monocyte/macrophage hematopoietic precursors that terminally adhere to mineralized extracellular matrix, and they subsequently break down the extracellular compartment. Bone is generally considered the load-burdening tissue, bone homeostasis is critically affected by mechanical conductions, and the bone cells are mechanosensitive. The functions of various bone cells under mechanical forces such as chondrocytes and osteoblasts have been reported; however, the unique bone-resorbing osteoclasts are less studied. The oversuppression of osteoclasts in mechanical studies may be because of its complicated differentiation progress and flexible structure, which increases difficulty in targeting mechanical structures. This paper will focus on recent findings regarding osteoclasts and attempt to uncover proposed candidate mechanosensing structures in osteoclasts including podosome-associated complexes, gap junctions and transient receptor potential family (ion channels). We will additionally describe possible mechanotransduction signaling pathways including GTPase ras homologue family member A (RhoA), Yes-associated protein/transcriptional co-activator with PDZ-binding motif (TAZ), Ca2+ signaling and non-canonical Wnt signaling. According to numerous studies, evaluating the possible influence of various physical environments on osteoclastogenesis is conducive to the study of bone homeostasis.
Collapse
Affiliation(s)
- Qingxuan Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Mengmeng Duan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Jingfeng Liao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
52
|
Sánchez JC, Ehrlich BE. Functional Interaction between Transient Receptor Potential V4 Channel and Neuronal Calcium Sensor 1 and the Effects of Paclitaxel. Mol Pharmacol 2021; 100:258-270. [PMID: 34321341 PMCID: PMC8626786 DOI: 10.1124/molpharm.121.000244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/08/2021] [Indexed: 11/22/2022] Open
Abstract
Neuronal calcium sensor 1 (NCS1), a calcium-binding protein, and transient receptor potential V4 (TRPV4), a plasma membrane calcium channel, are fundamental in the regulation of calcium homeostasis. The interactions of these proteins and their regulation by paclitaxel (PTX) were investigated using biochemical, pharmacological, and electrophysiological approaches in both a breast cancer epithelial cell model and a neuronal model. TRPV4 and NCS1 reciprocally immunoprecipitated each other, suggesting that they make up a signaling complex. The functional consequence of this physical association was that TRPV4 currents increased with increased NCS1 expression. Calcium fluxes through TRPV4 correlated with the magnitude of TRPV4 currents, and these calcium fluxes depended on NCS1 expression levels. Exposure to PTX amplified the acute effects of TRPV4 expression, currents, and calcium fluxes but decreased the expression of NCS1. These findings augment the understanding of the properties of TRPV4, the role of NCS1 in the regulation of TRPV4, and the cellular mechanisms of PTX-induced neuropathy. SIGNIFICANCE STATEMENT: TRPV4 and NCS1 physically and functionally interact. Increased expression of NCS1 enhances TRPV4-dependent currents, which are further amplified by treatment with the chemotherapeutic drug paclitaxel, an effect associated with adverse effects of chemotherapy, including neuropathy.
Collapse
Affiliation(s)
- Julio C Sánchez
- Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, Pereira, Colombia (J.C.S.), and Departments of Pharmacology and Cellular and Molecular Physiology, Yale University, New Haven, Connecticut (B.E.E.)
| | - Barbara E Ehrlich
- Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, Pereira, Colombia (J.C.S.), and Departments of Pharmacology and Cellular and Molecular Physiology, Yale University, New Haven, Connecticut (B.E.E.)
| |
Collapse
|
53
|
Fan X, Wang C, Han J, Ding X, Tang S, Ning L. Role of TRPV4-P2X7 Pathway in Neuropathic Pain in Rats with Chronic Compression of the Dorsal Root Ganglion. Neurochem Res 2021; 46:2143-2153. [PMID: 34014488 DOI: 10.1007/s11064-021-03352-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/16/2021] [Accepted: 05/12/2021] [Indexed: 10/21/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a Ca2+-permeable non-selective cation channel that is involved in the development of neuropathic pain. P2X7 receptor (P2X7) belongs to a class of ATP-gated nonselective cation channels that plays an important role in neuropathic pain. Nevertheless, little is known about the interaction between them for neuropathic pain. In this paper, we investigated role of TRPV4-P2X7 pathway in neuropathic pain. We evaluated the effect of TRPV4-P2X7 pathway on neuropathic pain in a chronic compression of the dorsal root ganglion (DRG) (hereafter termed CCD) model. We analyzed the effect of P2X7 on mechanical and thermal hyperalgesia mediated by TRPV4 in CCD. Furthermore, we assessed the effect of TRPV4 on the expression of P2X7 and the release of IL-1β and IL-6 in DRG after CCD. We found that intraperitoneal injection of TRPV4 agonist GSK-1016790A led to a significant increase of mechanical and thermal hyperalgesia in CCD, which was partially suppressed by P2X7 blockade with antagonist Brilliant Blue G (BBG). Then, we further noticed that GSK-1016790A injection increased the P2X7 expression of CCD, which was decreased by TRPV4 blockade with antagonist RN-1734 and HC-067047. Furthermore, we also discovered that the expressions of IL-1β and IL-6 were upregulated by GSK-1016790A injection but reduced by RN-1734 and HC-067047. Our results provide evidence that P2X7 contributes to development of neuropathic pain mediated by TRPV4 in the CCD model, which may be the basis for treatment of neuropathic pain relief.
Collapse
Affiliation(s)
- Xiaohua Fan
- Department of Rehabilitation Medicine, Shandong Provincial Hospital, 324 Jing Wu Wei Qi Road, Jinan, 250012, China
- Department of Rehabilitation Medicine, Shandong First Medical University, Taian, 250012, China
| | - Chuanwei Wang
- Department of Neurosurgery, Qilu Hospital Affiliated To Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Junting Han
- Department of Rehabilitation Medicine, Shandong First Medical University, Taian, 250012, China
| | - Xinli Ding
- Department of Rehabilitation Medicine, Shandong Provincial Hospital, 324 Jing Wu Wei Qi Road, Jinan, 250012, China
- Department of Rehabilitation Medicine, Shandong First Medical University, Taian, 250012, China
| | - Shaocan Tang
- Department of Rehabilitation Medicine, Shandong Provincial Hospital, 324 Jing Wu Wei Qi Road, Jinan, 250012, China
- Department of Rehabilitation Medicine, Shandong First Medical University, Taian, 250012, China
| | - Liping Ning
- Department of Rehabilitation Medicine, Shandong Provincial Hospital, 324 Jing Wu Wei Qi Road, Jinan, 250012, China.
- Department of Rehabilitation Medicine, Shandong First Medical University, Taian, 250012, China.
| |
Collapse
|
54
|
Tomilin VN, Pyrshev K, Khayyat NH, Zaika O, Pochynyuk O. With-No-Lysine Kinase 1 (WNK1) Augments TRPV4 Function in the Aldosterone-Sensitive Distal Nephron. Cells 2021; 10:1482. [PMID: 34204757 PMCID: PMC8231605 DOI: 10.3390/cells10061482] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 11/17/2022] Open
Abstract
Kidneys play a central role in regulation of potassium homeostasis and maintenance of plasma K+ levels within a narrow physiological range. With-no-lysine (WNK) kinases, specifically WNK1 and WNK4, have been recognized to regulate K+ balance, in part, by orchestrating maxi K+ channel (BK)-dependent K+ secretion in the aldosterone-sensitive distal nephron (ASDN), which includes the connecting tubule and collecting duct. We recently demonstrated that the Ca2+-permeable TRPV4 channel is essential for BK activation in the ASDN. Furthermore, high K+ diet increases TRPV4 activity and expression largely in an aldosterone-dependent manner. In the current study, we aimed to test whether WNK kinases contribute to regulation of TRPV4 activity and its stimulation by aldosterone. Systemic inhibition of WNK with WNK463 (1 mg/kgBW for 3 days) markedly decreased TRPV4-dependent Ca2+ influx in freshly isolated split-opened collecting ducts. Aldosterone greatly increased TRPV4 activity and expression in cultured mpkCCDc14 cells and this effect was abolished in the presence of WNK463. Selective inhibition of WNK1 with WNK-in-11 (400 nM, 24 h) recapitulated the effects of WNK463 on TRPV4-dependent Ca2+ influx. Interestingly, WNK-in-11 did not interfere with up-regulation of TRPV4 expression by aldosterone, but prevented translocation of the channel to the apical plasma membrane. Furthermore, co-expression of TRPV4 and WNK1 into Chinese hamster ovary (CHO) cells increased the macroscopic TRPV4-dependent cation currents. In contrast, over-expression of TRPV4 with a dominant negative WNK1 variant (K233M) decreased the whole-cell currents, suggesting both stimulatory and permissive roles of WNK1 in regulation of TRPV4 activity. Overall, we show that WNK1 is essential for setting functional TRPV4 expression in the ASDN at the baseline and in response to aldosterone. We propose that this new mechanism contributes to regulation of K+ secretion and, by extension, urinary K+ levels to maintain systemic potassium homeostasis.
Collapse
Affiliation(s)
| | | | | | | | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, 6431 Fannin, Houston, TX 77030, USA; (V.N.T.); (K.P.); (N.H.K.); (O.Z.)
| |
Collapse
|
55
|
Kawasaki S, Soga M, Sakurai Y, Nanchi I, Yamamoto M, Imai S, Takahashi T, Tsuno N, Asaki T, Morioka Y, Fujita M. Selective blockade of transient receptor potential vanilloid 4 reduces cyclophosphamide-induced bladder pain in mice. Eur J Pharmacol 2021; 899:174040. [PMID: 33737012 DOI: 10.1016/j.ejphar.2021.174040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/30/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a non-selective cation channel activated by various physical stimuli such as cell swelling and shear stress. TRPV4 is expressed in bladder sensory nerves and epithelium, and its activation produces urinary dysfunction in rodents. However, there have been few reports regarding its involvement in bladder pain. Therefore, we investigated whether TRPV4 is involved in bladder pain in mouse cystitis model. Intraperitoneal injection of cyclophosphamide (CYP; 300 mg/kg) produced mechanical hypersensitivity in the lower abdomen associated with a severe inflammatory bladder in mice. The mechanical threshold was reversed significantly in Trpv4-knockout (KO) mice. Repeated injections of CYP (150 mg/kg) daily for 4 days provoked mild bladder inflammation and persistent mechanical hypersensitivity in mice. Trpv4-KO mice prevented a reduction of the mechanical threshold without an alteration in bladder inflammation. A selective TRPV4 antagonist also reversed the mechanical threshold in chronic cystitis mice. Although expression of Trpv4 was unchanged in the bladders of chronic cystitis mice, the level of phosphorylated TRPV4 was increased significantly. These results suggest involvement of TRPV4 in bladder pain of cystitis mice. A TRPV4 antagonist might be useful for patients with irritable bladder pain such as those with interstitial cystitis/painful bladder syndrome.
Collapse
MESH Headings
- Analgesics/pharmacology
- Animals
- Behavior, Animal/drug effects
- Cells, Cultured
- Cyclophosphamide
- Cystitis, Interstitial/chemically induced
- Cystitis, Interstitial/metabolism
- Cystitis, Interstitial/physiopathology
- Cystitis, Interstitial/prevention & control
- Disease Models, Animal
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Nociceptive Pain/chemically induced
- Nociceptive Pain/metabolism
- Nociceptive Pain/physiopathology
- Nociceptive Pain/prevention & control
- Pain Threshold/drug effects
- Phosphorylation
- TRPV Cation Channels/antagonists & inhibitors
- TRPV Cation Channels/genetics
- TRPV Cation Channels/metabolism
- Urinary Bladder/drug effects
- Urinary Bladder/metabolism
- Urinary Bladder/physiopathology
- Mice
Collapse
Affiliation(s)
- Shiori Kawasaki
- Laboratory for Drug Discovery & Disease Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Masahiko Soga
- Animal Production Technology for Animal Models, Shionogi Techno Advance Research Co. Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Yusuke Sakurai
- Laboratory for Drug Discovery & Disease Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Isamu Nanchi
- Laboratory for Innovative Therapy Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Miyuki Yamamoto
- Laboratory for Drug Discovery & Disease Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Sunao Imai
- Laboratory for Advanced Medicine Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Tatsuya Takahashi
- Laboratory for Advanced Medicine Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Naoki Tsuno
- API R&D Laboratory, CMC R&D Division, Shionogi & Co., Ltd., 1-3, Kuise terajima 2-chome, Amagasaki, Hyogo, 660-0813, Japan
| | - Toshiyuki Asaki
- Laboratory for Drug Discovery & Disease Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Yasuhide Morioka
- Laboratory for Drug Discovery & Disease Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan
| | - Masahide Fujita
- Laboratory for Drug Discovery & Disease Research, Shionogi & Co., Ltd., 1-1 Futaba-cho 3-chome, Toyonaka, Osaka, 561-0825, Japan.
| |
Collapse
|
56
|
Ragamin A, Gomes CC, Bindels-de Heus K, Sandoval R, Bassenden AV, Dib L, Kok F, Alves J, Mathijssen I, Medici-Van den Herik E, Eveleigh R, Gayden T, Pullens B, Berghuis A, van Slegtenhorst M, Wilke M, Jabado N, Mancini GMS, Gomez RS. De novo TRPV4 Leu619Pro variant causes a new channelopathy characterised by giant cell lesions of the jaws and skull, skeletal abnormalities and polyneuropathy. J Med Genet 2021; 59:305-312. [PMID: 33685999 PMCID: PMC8867273 DOI: 10.1136/jmedgenet-2020-107427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/07/2020] [Accepted: 12/26/2020] [Indexed: 11/04/2022]
Abstract
BACKGROUND Pathogenic germline variants in Transient Receptor Potential Vanilloid 4 Cation Channel (TRPV4) lead to channelopathies, which are phenotypically diverse and heterogeneous disorders grossly divided in neuromuscular disorders and skeletal dysplasia. We recently reported in sporadic giant cell lesions of the jaws (GCLJs) novel, somatic, heterozygous, gain-of-function mutations in TRPV4, at Met713. METHODS Here we report two unrelated women with a de novo germline p.Leu619Pro TRPV4 variant and an overlapping systemic disorder affecting all organs individually described in TRPV4 channelopathies. RESULTS From an early age, both patients had several lesions of the nervous system including progressive polyneuropathy, and multiple aggressive giant cell-rich lesions of the jaws and craniofacial/skull bones, and other skeletal lesions. One patient had a relatively milder disease phenotype possibly due to postzygotic somatic mosaicism. Indeed, the TRPV4 p.Leu619Pro variant was present at a lower frequency (variant allele frequency (VAF)=21.6%) than expected for a heterozygous variant as seen in the other proband, and showed variable regional frequency in the GCLJ (VAF ranging from 42% to 10%). In silico structural analysis suggests that the gain-of-function p.Leu619Pro alters the ion channel activity leading to constitutive ion leakage. CONCLUSION Our findings define a novel polysystemic syndrome due to germline TRPV4 p.Leu619Pro and further extend the spectrum of TRPV4 channelopathies. They further highlight the convergence of TRPV4 mutations on different organ systems leading to complex phenotypes which are further mitigated by possible post-zygotic mosaicism. Treatment of this disorder is challenging, and surgical intervention of the GCLJ worsens the lesions, suggesting the future use of MEK inhibitors and TRPV4 antagonists as therapeutic modalities for unmet clinical needs.
Collapse
Affiliation(s)
- Aviel Ragamin
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Carolina C Gomes
- Department of Pathology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Karen Bindels-de Heus
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Renata Sandoval
- Oncogenetics, Hospital Sírio-Libanês, Brasília, Hospital Sirio-Libanes, Sao Paulo, Brazil
| | | | - Luciano Dib
- Post Graduation Program, School of Dentistry, Paulista University (UNIP), Sao Paulo, Brazil
| | - Fernando Kok
- Department of Neurology, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Julieta Alves
- Division of Neurosurgery, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Irene Mathijssen
- Department of Plastic and Reconstructive Surgery, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | - Robert Eveleigh
- Canadian Centre for Computational Genomics (C3G), Montreal, Québec, Canada.,McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Tenzin Gayden
- Department of Human Genetics, McGill University Faculty of Medicine, Montreal, Québec, Canada
| | - Bas Pullens
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC University Medical Center, Rotterdam, Zuid-Holland, The Netherlands
| | - Albert Berghuis
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Marjon van Slegtenhorst
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Martina Wilke
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Nada Jabado
- Department of Human Genetics, McGill University Faculty of Medicine, Montreal, Québec, Canada.,Department of Pediatrics, McGill University and McGill University Heath Centre Research Institute, Montreal, Quebec, Canada
| | - Grazia Maria Simonetta Mancini
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands .,ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ricardo Santiago Gomez
- Department of Oral Surgery and Pathology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
57
|
Neuropathy-causing TRPV4 mutations disrupt TRPV4-RhoA interactions and impair neurite extension. Nat Commun 2021; 12:1444. [PMID: 33664271 PMCID: PMC7933254 DOI: 10.1038/s41467-021-21699-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 02/02/2021] [Indexed: 12/19/2022] Open
Abstract
TRPV4 is a cell surface-expressed calcium-permeable cation channel that mediates cell-specific effects on cellular morphology and function. Dominant missense mutations of TRPV4 cause distinct, tissue-specific diseases, but the pathogenic mechanisms are unknown. Mutations causing peripheral neuropathy localize to the intracellular N-terminal domain whereas skeletal dysplasia mutations are in multiple domains. Using an unbiased screen, we identified the cytoskeletal remodeling GTPase RhoA as a TRPV4 interactor. TRPV4-RhoA binding occurs via the TRPV4 N-terminal domain, resulting in suppression of TRPV4 channel activity, inhibition of RhoA activation, and extension of neurites in vitro. Neuropathy but not skeletal dysplasia mutations disrupt TRPV4-RhoA binding and cytoskeletal outgrowth. However, inhibition of RhoA restores neurite length in vitro and in a fly model of TRPV4 neuropathy. Together these results identify RhoA as a critical mediator of TRPV4-induced cell structure changes and suggest that disruption of TRPV4-RhoA binding may contribute to tissue-specific toxicity of TRPV4 neuropathy mutations. TRPV4 dominant mutations cause neuropathy. Here, the authors show that TRPV4 binds and interacts with RhoA, modulating the actin cytoskeleton. Neuropathy-causing mutations of TRPV4 disrupt this complex, leading to RhoA activation and impairment of neurite extension in cultured cells and flies.
Collapse
|
58
|
Molecular modeling of three-dimensional structure of hTRPV4 protein and experimental verification of its antagonist binding sites. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
59
|
Vanneste M, Segal A, Voets T, Everaerts W. Transient receptor potential channels in sensory mechanisms of the lower urinary tract. Nat Rev Urol 2021; 18:139-159. [PMID: 33536636 DOI: 10.1038/s41585-021-00428-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 01/30/2023]
Abstract
Disruptions to sensory pathways in the lower urinary tract commonly occur and can give rise to lower urinary tract symptoms (LUTS). The unmet clinical need for treatment of LUTS has stimulated research into the molecular mechanisms that underlie neuronal control of the bladder and transient receptor potential (TRP) channels have emerged as key regulators of the sensory processes that regulate bladder function. TRP channels function as molecular sensors in urothelial cells and afferent nerve fibres and can be considered the origin of bladder sensations. TRP channels in the lower urinary tract contribute to the generation of normal and abnormal bladder sensations through a variety of mechanisms, and have demonstrated potential as targets for the treatment of LUTS in functional disorders of the lower urinary tract.
Collapse
Affiliation(s)
- Matthias Vanneste
- Laboratory of Ion Channel Research, VIB Center for Brain & Disease Research, Leuven, and Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Andrei Segal
- Laboratory of Ion Channel Research, VIB Center for Brain & Disease Research, Leuven, and Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, VIB Center for Brain & Disease Research, Leuven, and Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Wouter Everaerts
- Laboratory of Experimental Urology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
| |
Collapse
|
60
|
Henao JC, Grismaldo A, Barreto A, Rodríguez-Pardo VM, Mejía-Cruz CC, Leal-Garcia E, Pérez-Núñez R, Rojas P, Latorre R, Carvacho I, Torres YP. TRPM8 Channel Promotes the Osteogenic Differentiation in Human Bone Marrow Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:592946. [PMID: 33614639 PMCID: PMC7890257 DOI: 10.3389/fcell.2021.592946] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 01/05/2021] [Indexed: 11/29/2022] Open
Abstract
Various families of ion channels have been characterized in mesenchymal stem cells (MSCs), including some members of transient receptor potential (TRP) channels family. TRP channels are involved in critical cellular processes as differentiation and cell proliferation. Here, we analyzed the expression of TRPM8 channel in human bone marrow MSCs (hBM-MSCs), and its relation with osteogenic differentiation. Patch-clamp recordings showed that hBM-MSCs expressed outwardly rectifying currents which were increased by exposure to 500 μM menthol and were partially inhibited by 10 μM of BCTC, a TRPM8 channels antagonist. Additionally, we have found the expression of TRPM8 by RT-PCR and western blot. We also explored the TRPM8 localization in hBM-MSCs by immunofluorescence using confocal microscopy. Remarkably, hBM-MSCs treatment with 100 μM of menthol or 10 μM of icilin, TRPM8 agonists, increases osteogenic differentiation. Conversely, 20 μM of BCTC, induced a decrease of osteogenic differentiation. These results suggest that TRPM8 channels are functionally active in hBM-MSCs and have a role in cell differentiation.
Collapse
Affiliation(s)
- Juan C Henao
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Adriana Grismaldo
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Alfonso Barreto
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Viviana M Rodríguez-Pardo
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Claudia Camila Mejía-Cruz
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Efrain Leal-Garcia
- Departamento de Ortopedia y Traumatología, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Patricio Rojas
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Ramón Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ingrid Carvacho
- Department of Biology and Chemistry, Faculty of Basic Sciences, Universidad Católica del Maule, Talca, Chile
| | - Yolima P Torres
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
61
|
TRPing to the Point of Clarity: Understanding the Function of the Complex TRPV4 Ion Channel. Cells 2021; 10:cells10010165. [PMID: 33467654 PMCID: PMC7830798 DOI: 10.3390/cells10010165] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
The transient receptor potential vanilloid 4 channel (TRPV4) belongs to the mammalian TRP superfamily of cation channels. TRPV4 is ubiquitously expressed, activated by a disparate array of stimuli, interacts with a multitude of proteins, and is modulated by a range of post-translational modifications, the majority of which we are only just beginning to understand. Not surprisingly, a great number of physiological roles have emerged for TRPV4, as have various disease states that are attributable to the absence, or abnormal functioning, of this ion channel. This review will highlight structural features of TRPV4, endogenous and exogenous activators of the channel, and discuss the reported roles of TRPV4 in health and disease.
Collapse
|
62
|
Liu Y, Yan X, Chen Y, He Z, Ouyang Y. Novel TRPV4 mutation in a large Chinese family with congenital distal spinal muscular atrophy, skeletal dysplasia and scaly skin. J Neurol Sci 2020; 419:117153. [DOI: 10.1016/j.jns.2020.117153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 12/28/2022]
|
63
|
|
64
|
Sun L, Yao K, Zhang H, Chen W. Activation of the ATP-P2X pathway by TRPV4 in acute ocular hypertension. Int J Ophthalmol 2020; 13:1697-1704. [PMID: 33214998 DOI: 10.18240/ijo.2020.11.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 08/06/2020] [Indexed: 11/23/2022] Open
Abstract
AIM To measure the expression of transient receptor potential cation channel subfamily V member 4 (TRPV4) in the rat cornea and determine whether it is related to adenosine triphosphate (ATP) generation in a rat model of acute ocular hypertension (AOH). METHODS Immunofluorescence staining of TRPV4, P2X2 receptor, P2X3 receptor, and β3-tubulin in rat corneal longitudinal sections and paved was performed to clearly display histological structures. Rat models of AOH and agonist/antagonist-treated groups were established and corneal ATP was measured using an ATP assay. The independent t-test and simple linear correlation model were adopted for statistical analyses. RESULTS Immunofluorescence staining of rat cornea sections revealed that epithelial and endothelial membranes showed strong immunoreactivity for TRPV4 and P2X2 receptor and coexpression with β3-tubulin in the rat corneal epithelial layer. Corneal ATP was significantly higher in the AOH rat model than in the control (P<0.05) and apparently lower after pretreatment by applying eyedrops of TRPV4 antagonist RN1734 with 30-40 mm Hg intraocular pressure (IOP; P<0.05). A simple linear regression model showed a positive correlation between rat corneal ATP and IOP values (R 2=0.996, P=0.0134) from the normal IOP (113 mm Hg) to 40 mm Hg. At 10-40min after anterior chamber injection of GSK1016790A (0.01 mL, 50 nmol/L in 0.9% NaCl), corneal ATP was significantly higher than in the control group (P<0.05), which peaked at 10min. The ATP concentration of the normal epithelium was higher than that of the endothelium in the AOH rat model and after anterior chamber injection of GSK1016790A (P<0.05). CONCLUSION The ATP concentration in the AOH rat cornea is increased by TRPV4 activation.
Collapse
Affiliation(s)
- Li Sun
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, Hubei Province, China
| | - Ke Yao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, Hubei Province, China
| | - Hong Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, Hubei Province, China
| | - Wei Chen
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, Hubei Province, China
| |
Collapse
|
65
|
Achanta S, Jordt SE. Transient receptor potential channels in pulmonary chemical injuries and as countermeasure targets. Ann N Y Acad Sci 2020; 1480:73-103. [PMID: 32892378 PMCID: PMC7933981 DOI: 10.1111/nyas.14472] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/22/2020] [Accepted: 07/29/2020] [Indexed: 12/17/2022]
Abstract
The lung is highly sensitive to chemical injuries caused by exposure to threat agents in industrial or transportation accidents, occupational exposures, or deliberate use as weapons of mass destruction (WMD). There are no antidotes for the majority of the chemical threat agents and toxic inhalation hazards despite their use as WMDs for more than a century. Among several putative targets, evidence for transient receptor potential (TRP) ion channels as mediators of injury by various inhalational chemical threat agents is emerging. TRP channels are expressed in the respiratory system and are essential for homeostasis. Among TRP channels, the body of literature supporting essential roles for TRPA1, TRPV1, and TRPV4 in pulmonary chemical injuries is abundant. TRP channels mediate their function through sensory neuronal and nonneuronal pathways. TRP channels play a crucial role in complex pulmonary pathophysiologic events including, but not limited to, increased intracellular calcium levels, signal transduction, recruitment of proinflammatory cells, neurogenic inflammatory pathways, cough reflex, hampered mucus clearance, disruption of the integrity of the epithelia, pulmonary edema, and fibrosis. In this review, we summarize the role of TRP channels in chemical threat agents-induced pulmonary injuries and how these channels may serve as medical countermeasure targets for broader indications.
Collapse
Affiliation(s)
- Satyanarayana Achanta
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina
| | - Sven-Eric Jordt
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
66
|
Sánchez JC, Muñoz LV, Ehrlich BE. Modulating TRPV4 channels with paclitaxel and lithium. Cell Calcium 2020; 91:102266. [PMID: 32871457 DOI: 10.1016/j.ceca.2020.102266] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/16/2020] [Accepted: 08/06/2020] [Indexed: 12/18/2022]
Abstract
Transient receptor potential V4 (TRPV4), a plasma membrane calcium channel, is implicated as a contributor to the initiation of chemotherapy-induced peripheral neuropathy (CIPN). Paclitaxel (PTX) is a commonly used anticancer drug that causes CIPN and lithium has been shown to prevent CIPN. However, the direct effect of PTX and lithium on TRPV4 is not clear. This study investigated these actions using biochemical, pharmacological, and electrophysiological approaches using a neuronal cell line (SH-SY5Y). The addition of pharmacologically appropriate levels of PTX increased the expression of TRPV4, TRPV4 currents, and TRPV4-dependent calcium fluxes. Prolonged exposure to PTX amplified the acute effects of TRPV4 expression, currents, and calcium fluxes. Pretreatment with lithium (1 mM) decreased TRPV4 currents and calcium fluxes in the absence and presence of PTX. These findings enhance our understanding of the properties and regulation of TRPV4, the cellular mechanisms of PTX-induced neuropathy, and the mechanism of lithium for prevention of CIPN.
Collapse
Affiliation(s)
- Julio C Sánchez
- Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, Pereira, 660003, Colombia.
| | - Laura V Muñoz
- Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, Pereira, 660003, Colombia
| | - Barbara E Ehrlich
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
67
|
Guarino BD, Paruchuri S, Thodeti CK. The role of TRPV4 channels in ocular function and pathologies. Exp Eye Res 2020; 201:108257. [PMID: 32979394 DOI: 10.1016/j.exer.2020.108257] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022]
Abstract
Transient potential receptor vanilloid 4 (TRPV4) is an ion channel responsible for sensing osmotic and mechanical signals, which in turn regulates calcium signaling across cell membranes. TRPV4 is widely expressed throughout the body, and plays an important role in normal physiological function, as well as different pathologies, however, its role in the eye is not well known. In the eye, TRPV4 is expressed in various tissues, such as the retina, corneal epithelium, ciliary body, and the lens. In this review, we provide an overview on TRPV4 structure, activation, mutations, and summarize the current knowledge of TRPV4 function and signaling mechanisms in various locations throughout the eye, as well as its role in ocular diseases, such as glaucoma and diabetic retinopathy. Based on the available data, we highlight the therapeutic potential of TRPV4 as well as the shortcomings of current research. Finally, we provide future perspectives on the implications of targeting TRPV4 to treat various ocular pathologies.
Collapse
Affiliation(s)
- Brianna D Guarino
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | | | - Charles K Thodeti
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA.
| |
Collapse
|
68
|
Fernández-Carvajal A, González-Muñiz R, Fernández-Ballester G, Ferrer-Montiel A. Investigational drugs in early phase clinical trials targeting thermotransient receptor potential (thermoTRP) channels. Expert Opin Investig Drugs 2020; 29:1209-1222. [PMID: 32941080 DOI: 10.1080/13543784.2020.1825680] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Thermo transient receptor potential (thermoTRP) channels are some of the most intensely pursued therapeutic targets of the past decade. They are considered promising targets of numerous diseases including chronic pain and cancer. Modulators of these proteins, in particular TRPV1-4, TRPM8 and TRPA1, have reached clinical development, but none has been approved for clinical practice yet. AREAS COVERED The therapeutic potential of targeting thermoTRP channels is discussed. The discussion is centered on our experience and on available data found in SciFinder, PubMed, and ClinicalTrials.gov database from the past decade. This review focuses on the therapeutic progress concerning this family of channels, including strategies to improve their therapeutic index for overcoming adverse effects. EXPERT OPINION Although thermoTRPs are pivotal drug targets, translation to the clinic has faced two key problems, (i) unforeseen side effects in Phase I trials and, (ii) poor clinical efficacy in Phase II trials. Thus, there is a need for (i) an enhanced understanding of the physiological role of these channels in tissues and organs and (ii) the development of human-based pre-clinical models with higher clinical translation. Furthermore, progress in nanotechnology-based delivery strategies will positively impact thermoTRP human pharmacology.
Collapse
Affiliation(s)
- Asia Fernández-Carvajal
- Instituto De Investigación, Desarrollo E Innovación En Biotecnología Sanitaria De Elche (Idibe), Universitas Miguel Hernández , Alicante, Spain
| | | | - Gregorio Fernández-Ballester
- Instituto De Investigación, Desarrollo E Innovación En Biotecnología Sanitaria De Elche (Idibe), Universitas Miguel Hernández , Alicante, Spain
| | - Antonio Ferrer-Montiel
- Instituto De Investigación, Desarrollo E Innovación En Biotecnología Sanitaria De Elche (Idibe), Universitas Miguel Hernández , Alicante, Spain
| |
Collapse
|
69
|
Zhang X, Mao Z, Huang Y, Zhang Z, Yao J. Gap junctions amplify TRPV4 activation-initiated cell injury via modification of intracellular Ca 2+ and Ca 2+-dependent regulation of TXNIP. Channels (Austin) 2020; 14:246-256. [PMID: 32752916 PMCID: PMC7515575 DOI: 10.1080/19336950.2020.1803552] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The elevated intracellular Ca2+ and oxidative stress are well-reported mechanisms behind renal tubular epithelial injury initiated by various insults. Given that TRPV4 and connexin43 (Cx43) channels are activated by a wide range of stimuli and regulate both intracellular Ca2+ and redox status, we speculated an involvement of these channels in renal tubular cell injury. Here, we tested this possibility and explored the potential underlying mechanisms. Our results demonstrated that exposure of renal tubular epithelial cells to aminoglycoside G418 led to cell death, which was attenuated by both TRPV4 and gap junction (Gj) inhibitor. Activation of TRPV4 caused cell damage, which was associated with an early increase in Cx43 expression and function. Inhibition of Cx43 with chemical inhibitor or siRNA largely prevented TRPV4 activation-induced cell damage. Further analysis revealed that TRPV4 agonists elicited a rise in intracellular Ca2+ and caused a Ca2+-dependent elevation in TXNIP (a negative regulator of the antioxidant thioredoxin). In the presence of Gj inhibitor, however, these effects of TRPV4 were largely prevented. The depletion of intracellular Ca2+ with Ca2+ chelator BAPTA-AM or downregulation of TXNIP with siRNA significantly alleviated TRPV4 activation-initiated cell injury. Collectively, our results point to a critical involvement of TRPV4/Cx43 channel interaction in renal tubular cell injury through mechanisms involving a synergetic induction of intracellular Ca2+ and oxidative stress. Channel interactions could be an important mechanism underlying cell injury. Targeting channels could have therapeutic potential for the treatment of acute tubular cell injury.
Collapse
Affiliation(s)
- Xiling Zhang
- Department of Urology, The Fourth Affiliated Hospital of China Medical University , Shenyang, China.,Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi , Chuo, Japan
| | - Zhimin Mao
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi , Chuo, Japan
| | - Yanru Huang
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi , Chuo, Japan
| | - Zhen Zhang
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi , Chuo, Japan
| | - Jian Yao
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi , Chuo, Japan
| |
Collapse
|
70
|
Sharma S, Ma L, Rahaman SO. Role of TRPV4 in matrix stiffness-induced expression of EMT-specific LncRNA. Mol Cell Biochem 2020; 474:189-197. [PMID: 32734537 DOI: 10.1007/s11010-020-03844-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/17/2020] [Indexed: 02/06/2023]
Abstract
Long non-coding RNAs (LncRNAs) are long (> 200 bases), non-coding, single-stranded RNAs that have emerged as major regulators of gene expression, cell differentiation, development, and oncogenesis. In view of the fact that matrix stiffness plays a role in cellular functions associated with these processes, it is important to ask what role matrix stiffness plays in regulating expression of LncRNAs. In this report, we show that (i) matrix stiffness causes differential expression of epithelial-mesenchymal transition (EMT)-related LncRNAs and mRNAs in primary mouse normal epidermal keratinocytes, (ii) differential expression of EMT-related LncRNAs and mRNAs occurs in response to combined stimulation of transforming growth factor β1 and matrix stiffness, and (iii) transient receptor potential (TRP) channel of the vanilloid subfamily, TRPV4, a matrix stiffness-sensitive ion channel, plays a role in differential expression of EMT-related LncRNAs and mRNAs in response to combined stimulation by TGFβ1 and matrix stiffness. These data identify TRPV4 as a candidate plasma membrane mechanosensor that transmits matrix-sensing signals essential to LncRNA expression. Our results also show that we have established and validated an assay system capable of discovering novel LncRNAs and mRNAs sensitive to matrix stiffening.
Collapse
Affiliation(s)
- Shweta Sharma
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, 20742, USA
| | - Li Ma
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Shaik O Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
71
|
The intracellular calcium dynamics in a single vascular endothelial cell being squeezed through a narrow microfluidic channel. Biomech Model Mechanobiol 2020; 20:55-67. [PMID: 32710185 DOI: 10.1007/s10237-020-01368-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/11/2020] [Indexed: 12/17/2022]
Abstract
Revealing the mechanisms underlying the intracellular calcium responses in vascular endothelial cells (VECs) induced by mechanical stimuli contributes to a better understanding for vascular diseases, including hypertension, atherosclerosis, and aneurysm. Combining with experimental measurement and Computational Fluid Dynamics simulation, we developed a mechanobiological model to investigate the intracellular [Ca2+] response in a single VEC being squeezed through narrow microfluidic channel. The time-dependent cellular surface tension dynamics was quantified throughout the squeezing process. In our model, the various Ca2+ signaling pathways activated by mechanical stimulation is fully considered. The simulation results of our model exhibited well agreement with our experimental results. By using the model, we theoretically explored the mechanism of the two-peak intracellular [Ca2+] response in single VEC being squeezed through narrow channel and made some testable predictions for guiding experiment in the future.
Collapse
|
72
|
Boudaka A, Saito CT, Tominaga M. Deletion of TRPV4 enhances in vitro wound healing of murine esophageal keratinocytes. Sci Rep 2020; 10:11349. [PMID: 32647282 PMCID: PMC7347589 DOI: 10.1038/s41598-020-68269-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/22/2020] [Indexed: 12/21/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a non-selective cation channel that is widely expressed in different body tissues and plays several physiological roles. This channel is highly expressed in esophageal keratinocytes where its activation mediates ATP release. However, whether TRPV4 has a role in wound healing of esophageal keratinocytes is unclear. In this study, we demonstrated that both cell migration and proliferation were slower in wild-type esophageal keratinocytes compared to cells having TRPV4 knockout. Our results suggest that TRPV4-mediated release of ATP from esophageal keratinocytes contributes to a decrease in the rate of in vitro wound healing via the ATP degradation product adenosine, which acts on A2B adenosine receptors.
Collapse
Affiliation(s)
- Ammar Boudaka
- Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Al-Khoud, P.O. Box 35, 123, Muscat, Sultanate of Oman.
- Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Aichi, 444-8787, Japan.
| | - Claire T Saito
- Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Aichi, 444-8787, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, Aichi, 444-8787, Japan
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, 444-8787, Japan
- Thermal Biology Group, Exploratory Research Center on Life and Living Systems, Okazaki, Aichi, 444-8787, Japan
| |
Collapse
|
73
|
Scheraga RG, Southern BD, Grove LM, Olman MA. The Role of TRPV4 in Regulating Innate Immune Cell Function in Lung Inflammation. Front Immunol 2020; 11:1211. [PMID: 32676078 PMCID: PMC7333351 DOI: 10.3389/fimmu.2020.01211] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/15/2020] [Indexed: 12/22/2022] Open
Abstract
Ion channels/pumps are essential regulators of innate immune cell function. Macrophages have been increasingly recognized to have phenotypic plasticity and location-specific functions in the lung. Transient receptor potential vanilloid 4 (TRPV4) function in lung injury has been shown to be stimulus- and cell-type specific. In the current review, we discuss the importance of TRPV4 in macrophages and its role in phagocytosis and cytokine secretion in acute lung injury/acute respiratory distress syndrome (ARDS). Furthermore, TRPV4 controls a MAPK molecular switch from predominately c-Jun N-terminal kinase, JNK activation, to that of p38 activation, that mediates phagocytosis and cytokine secretion in a matrix stiffness-dependent manner. Expanding knowledge regarding the downstream mechanisms by which TRPV4 acts to tailor macrophage function in pulmonary inflammatory diseases will allow for formulation of novel therapeutics.
Collapse
Affiliation(s)
- Rachel G. Scheraga
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Brian D. Southern
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Lisa M. Grove
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Mitchell A. Olman
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
74
|
Zhang N, Xing Y, Yu Y, Liu C, Jin B, Huo L, Kong D, Yang Z, Zhang X, Zheng R, Jia Z, Kang L, Zhang W. Influence of human amylin on the membrane stability of rat primary hippocampal neurons. Aging (Albany NY) 2020; 12:8923-8938. [PMID: 32463790 PMCID: PMC7288967 DOI: 10.18632/aging.103105] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/09/2020] [Indexed: 04/09/2023]
Abstract
The two most common aging-related diseases, Alzheimer's disease and type 2 diabetes mellitus, are associated with accumulation of amyloid proteins (β-amyloid and amylin, respectively). This amylin aggregation is reportedly cytotoxic to neurons. We found that aggregation of human amylin (hAmylin) induced neuronal apoptosis without obvious microglial infiltration in vivo. High concentrations of hAmylin irreversibly aggregated on the surface of the neuronal plasma membrane. Long-term incubation with hAmylin induced morphological changes in neurons. Moreover, hAmylin permeabilized the neuronal membrane within 1 min in a manner similar to Triton X-100, allowing impermeable fluorescent antibodies to enter the neurons and stain intracellular antigens. hAmylin also permeabilized the cell membrane of astrocytes, though more slowly. Under scanning electron microscopy, we observed that hAmylin destroyed the integrity of the cell membranes of both neurons and astrocytes. Additionally, it increased intracellular reactive oxygen species generation and reduced the mitochondrial membrane potential. Thus, by aggregating on the surface of neurons, hAmylin impaired the cell membrane integrity, induced reactive oxygen species production, reduced the mitochondrial membrane potential, and ultimately induced neuronal apoptosis.
Collapse
Affiliation(s)
- Nan Zhang
- Central Laboratory, First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei International Joint Research Center for Brain Science, Shijiazhuang, Hebei, China
| | - Yuan Xing
- Department of Neurology, First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Yongzhou Yu
- Department of Pharmacology, Institution of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chao Liu
- Department of Laboratory Animal Science, Hebei Medical University, Hebei Key Lab of Laboratory Animal Science, Shijiazhuang, Hebei, China
| | - Baohua Jin
- Department of Pharmacology, Institution of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lifang Huo
- Department of Pharmacology, Institution of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dezhi Kong
- Department of Pharmacology, Institution of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zuxiao Yang
- Department of Pharmacology, Institution of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiangjian Zhang
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, Health Science Center, Neuroscience Research Institute, Key Laboratory for Neuroscience of the Ministry of Education, Key Laboratory for Neuroscience of the National Health Commission, Peking University, Beijing, China
| | - Zhanfeng Jia
- Department of Pharmacology, The Key Laboratory of New Drug Pharmacology and Toxicology, Center for Innovative Drug Research and Evaluation, Institute of Medical Science and Health, The Key Laboratory of Neural and Vascular Biology Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lin Kang
- Department of Endocrinology, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Clinical Medical Research Center, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Wei Zhang
- Department of Pharmacology, Institution of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
75
|
Alpizar YA, Uvin P, Naert R, Franken J, Pinto S, Sanchez A, Gevaert T, Everaerts W, Voets T, De Ridder D, Talavera K. TRPV4 Mediates Acute Bladder Responses to Bacterial Lipopolysaccharides. Front Immunol 2020; 11:799. [PMID: 32435246 PMCID: PMC7218059 DOI: 10.3389/fimmu.2020.00799] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/07/2020] [Indexed: 12/24/2022] Open
Abstract
Urinary tract infections (UTI) affect a large proportion of the population, causing among other symptoms, more frequent and urgent micturition. Previous studies reported that the gram-negative bacterial wall component lipopolysaccharides (LPS) trigger acute epithelial and bladder voiding responses, but the underlying mechanisms remain unknown. The cation channel TRPV4 is implicated in the regulation of the bladder voiding. Since TRPV4 is activated by LPS in airway epithelial cells, we sought to determine whether this channel plays a role in LPS-induced responses in urothelial cells (UCs). We found that human-derived UCs display a fast increase in intracellular Ca2+ concentration upon acute application of Escherichia coli LPS. Such responses were detected also in freshly isolated mouse UCs, and found to be dependent on TRPV4, but not to require the canonical TLR4 signaling pathway of LPS detection. Confocal microscopy experiments revealed that TRPV4 is dispensable for LPS-induced nuclear translocation of NF-κB in mouse UCs. On the other hand, quantitative RT PCR determinations showed an enhanced LPS-induced production of proinflammatory cytokines in TRPV4-deficient UCs. Cystometry experiments in anesthetized wild type mice revealed that acute intravesical instillation of LPS rapidly increases voiding frequency. This effect was not observed in TRPV4-deficient animals, but was largely preserved in Tlr4 KO and Trpa1 KO mice. Our results suggest that activation of TRPV4 by LPS in UCs regulates the proinflammatory response and contributes to LPS-induced increase in voiding frequency. These findings further support the concept that TRP channels are sensors of LPS, mediating fast innate immunity mechanisms against gram-negative bacteria.
Collapse
Affiliation(s)
- Yeranddy A Alpizar
- Laboratory for Ion Channel Research, Department of Cellular and Molecular Medicine, VIB Center for Brain & Disease Research, Leuven, Belgium.,VIB Center for Brain & Disease Research, Leuven, Belgium
| | - Pieter Uvin
- Laboratory for Ion Channel Research, Department of Cellular and Molecular Medicine, VIB Center for Brain & Disease Research, Leuven, Belgium.,Laboratory of Organ System, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Robbe Naert
- Laboratory for Ion Channel Research, Department of Cellular and Molecular Medicine, VIB Center for Brain & Disease Research, Leuven, Belgium.,VIB Center for Brain & Disease Research, Leuven, Belgium
| | - Jan Franken
- Laboratory for Ion Channel Research, Department of Cellular and Molecular Medicine, VIB Center for Brain & Disease Research, Leuven, Belgium.,Laboratory of Organ System, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Silvia Pinto
- Laboratory for Ion Channel Research, Department of Cellular and Molecular Medicine, VIB Center for Brain & Disease Research, Leuven, Belgium.,VIB Center for Brain & Disease Research, Leuven, Belgium
| | - Alicia Sanchez
- Laboratory for Ion Channel Research, Department of Cellular and Molecular Medicine, VIB Center for Brain & Disease Research, Leuven, Belgium.,VIB Center for Brain & Disease Research, Leuven, Belgium
| | - Thomas Gevaert
- Laboratory of Organ System, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Wouter Everaerts
- Laboratory for Ion Channel Research, Department of Cellular and Molecular Medicine, VIB Center for Brain & Disease Research, Leuven, Belgium.,Laboratory of Organ System, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Thomas Voets
- Laboratory for Ion Channel Research, Department of Cellular and Molecular Medicine, VIB Center for Brain & Disease Research, Leuven, Belgium.,VIB Center for Brain & Disease Research, Leuven, Belgium
| | - Dirk De Ridder
- Laboratory of Organ System, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Karel Talavera
- Laboratory for Ion Channel Research, Department of Cellular and Molecular Medicine, VIB Center for Brain & Disease Research, Leuven, Belgium.,VIB Center for Brain & Disease Research, Leuven, Belgium
| |
Collapse
|
76
|
Gupta N, Goswami R, Alharbi MO, Biswas D, Rahaman SO. TRPV4 is a regulator in P. gingivalis lipopolysaccharide-induced exacerbation of macrophage foam cell formation. Physiol Rep 2020; 7:e14069. [PMID: 30980509 PMCID: PMC6461712 DOI: 10.14814/phy2.14069] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/21/2019] [Accepted: 03/25/2019] [Indexed: 01/21/2023] Open
Abstract
Porphyromonas gingivalis (P.g), a major causative agent of periodontitis, has been linked to atherosclerosis, a chronic inflammatory vascular disease. Recent studies have suggested a link between periodontitis and arterial stiffness, a risk factor for atherosclerosis. However, the mechanisms by which P.g infection contributes to atherogenesis remain elusive. The formation of lipid-laden macrophage "foam cells" is critically important to development and progression of atherosclerosis. We have obtained evidence that TRPV4 (transient receptor potential channel of the vanilloid subfamily 4), a mechanosensitive channel, is a regulator of macrophage foam cell formation both in response to P.g-derived lipopolysaccharide (PgLPS) or to an increase in matrix stiffness. Importantly, we found that TRPV4 activity (Ca2+ influx) was increased in response to PgLPS. Genetic deletion or chemical antagonism of TRPV4 channels blocked PgLPS-triggered exacerbation of oxidized LDL (oxLDL)-mediated foam cell formation. Mechanistically, we found that (1) TRPV4 regulated oxLDL uptake but not its cell surface binding in macrophages; (2) reduced foam cell formation in TRPV4 null cells was independent of expression of CD36, a predominant receptor for oxLDL, and (3) co-localization of TRPV4 and CD36 on the macrophage plasma membrane was sensitive to the increased level of matrix stiffness occurring in the presence of PgLPS. Altogether, our results suggest that TRPV4 channels play an essential role in P.g-induced exacerbation of macrophage foam cell generation through a mechanism that modulates uptake of oxLDL.
Collapse
Affiliation(s)
- Nabyendu Gupta
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| | - Rishov Goswami
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| | - Mazen O Alharbi
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| | - Debabrata Biswas
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Shaik O Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| |
Collapse
|
77
|
Kim JH, Kwon TR, Lee SE, Jang YN, Han HS, Mun SK, Kim BJ. Comparative Evaluation of the Effectiveness of Novel Hyaluronic Acid-Polynucleotide Complex Dermal Filler. Sci Rep 2020; 10:5127. [PMID: 32198443 PMCID: PMC7083941 DOI: 10.1038/s41598-020-61952-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/18/2020] [Indexed: 11/08/2022] Open
Abstract
HA (Hyaluronic acid) filler, the most commonly used dermal filler, causes several side effects. HA-PN (Hyaluronic acid-Polynucleotide), a new composite filler, has excellent biocompatibility and induces tissue regeneration. In this study, we compare the efficacies and safety profiles of these fillers. The characteristics of HA and HA-PN fillers were compared using scanning electron microscopy and rheometry. No morphological difference was noted between the fillers. However, the latter had higher viscosity and elasticity values. The HA-PN filler induced higher cell migration than the HA filler in a wound healing assay. It was also found to stimulate better collagen synthesis in human and mouse fibroblasts. The HA and HA-PN fillers were injected into SKH1 hairless mice to determine changes in their volume for up to 24 weeks. Increased cell migration and collagen synthesis were observed in mice injected with the HA-PN complex filler. Although the safety and durability of the HA and HA-PN fillers were similar, the latter induced a lower transient receptor potential vanilloid 4 expression and caused less stimulation upon injection. In conclusion, HA-PN complex fillers can stimulate fibroblast growth and facilitate volume growth and skin regeneration.
Collapse
Affiliation(s)
- Jong Hwan Kim
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Tae-Rin Kwon
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Sung Eun Lee
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
- Department of Medicine, Graduate School, Chung-Ang University, Seoul, Korea
| | - Yoo Na Jang
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
- Department of Medicine, Graduate School, Chung-Ang University, Seoul, Korea
| | - Hye Sung Han
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
- Department of Medicine, Graduate School, Chung-Ang University, Seoul, Korea
| | - Seog Kyun Mun
- Department of Otorhinolaryngology-Head and Neck Surgery, Chung-Ang University College of Medicine, Seoul, Korea
| | - Beom Joon Kim
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea.
- Department of Medicine, Graduate School, Chung-Ang University, Seoul, Korea.
| |
Collapse
|
78
|
Role of macrophage TRPV4 in inflammation. J Transl Med 2020; 100:178-185. [PMID: 31645630 PMCID: PMC7261496 DOI: 10.1038/s41374-019-0334-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/19/2019] [Accepted: 08/30/2019] [Indexed: 01/05/2023] Open
Abstract
Transient receptor ion channels have emerged as immensely important channels/receptors in diverse physiological and pathological responses. Of particular interest is the transient receptor potential channel subfamily V member 4 (TRPV4), which is a polymodal, nonselective, calcium-permeant cation channel, and is activated by both endogenous and exogenous stimuli. Both neuronal and nonneuronal cells express functional TRPV4, which is responsive to a variety of biochemical and biomechanical stimuli. Emerging discoveries have advanced our understanding of the role of macrophage TRPV4 in numerous inflammatory diseases. In lung injury, TRPV4 mediates macrophage phagocytosis, secretion of pro-resolution cytokines, and generation of reactive oxygen species. TRPV4 regulates lipid-laden macrophage foam cell formation, the hallmark of atheroinflammatory conditions, in response to matrix stiffness and lipopolysaccharide stimulation. Accumulating data also point to a role of macrophage TRPV4 in the pathogenesis of the foreign body response, a chronic inflammatory condition, through the formation of foreign body giant cells. Deletion of TRPV4 in macrophages suppresses the allergic and nonallergic itch in a mouse model, suggesting a role of TRPV4 in skin disease. Here, we discuss the current understanding of the role of macrophage TRPV4 in various inflammatory conditions.
Collapse
|
79
|
TRPV4 is dispensable for the development of airway allergic asthma. J Transl Med 2020; 100:265-273. [PMID: 31417159 PMCID: PMC7261591 DOI: 10.1038/s41374-019-0305-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 07/17/2019] [Accepted: 07/23/2019] [Indexed: 11/08/2022] Open
Abstract
Allergic asthma is one of the most common immune-mediated disorders affecting the lungs. It is characterized clinically by airway hyperresponsiveness, eosinophilia, enhanced IL-4 and IL-13, peribronchial inflammation with mononuclear cell infiltration, and goblet cell hyperplasia associated with increased mucus production. However, chronic asthma with repeated exposures to inhaled allergens can result in subepithelial pulmonary fibrosis. The transient receptor potential cation channel subfamily V member 4 (TRPV4) protein can promote the generation of myofibroblasts and pulmonary fibrosis. Here, we investigated the possibility that TPRV4 facilitates the development of allergic asthma and subsequent pulmonary fibrosis in the lung. To test this, wild-type (WT) and TPRV4 gene knockout (KO) mice were repeatedly sensitized with chicken ovalbumin (OVA) and repeatedly subjected to aerosol challenge with 1% OVA. We found that there were no significant differences in the development of allergic asthma between the WT and TPRV4 KO mice. Both groups of mice exhibited similar levels of airway hyperresponsiveness, IL-13, IL-5, OVA-specific IgE, eosinophilia, mucus-secreting goblet cell hyperplasia, and deposition of collagen fiber, which is a hallmark of the pulmonary fibrosis. Thus, these data suggest that TPRV4 protein is dispensable in the initiation and development of airway asthma and subsequent fibrosis.
Collapse
|
80
|
Cytochrome P450 Epoxygenase-Dependent Activation of TRPV4 Channel Participates in Enhanced Serotonin-Induced Pulmonary Vasoconstriction in Chronic Hypoxic Pulmonary Hypertension. Anal Cell Pathol (Amst) 2020; 2020:8927381. [PMID: 32399392 PMCID: PMC7204149 DOI: 10.1155/2020/8927381] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 12/10/2019] [Accepted: 12/28/2019] [Indexed: 12/26/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a multi-functional non-selective channel expressed in pulmonary vasculatures. TRPV4 contributes to serotonin- (5-HT-) induced pulmonary vasoconstriction and is responsible in part for the enhanced 5-HT response in pulmonary arteries (PAs) of chronic hypoxia mice. Epoxyeicosatrienoic acid (EET) is an endogenous agonist of TRPV4 and is known to regulate vasoreactivity. The levels of EETs, the expression of cytochrome P450 (CYP) epoxygenase for EET production, and epoxide hydrolase for EET degradation are altered by chronic hypoxia. Here, we examined the role of EET-dependent TRPV4 activation in the 5-HT-mediated PA contraction. In PAs of normoxic mice, inhibition of TRPV4 with a specific inhibitor HC-067047 caused a decrease in the sensitivity of 5-HT-induced PA contraction without affecting the maximal contractile response. Application of the cytochrome P450 epoxygenase inhibitor MS-PPOH had no effect on the vasoreactivity to 5-HT. In contrast, inhibition of CYP epoxygenase or TRPV4 both attenuated the 5-HT-elicited maximal contraction to a comparable level in PAs of chronic hypoxic mice. Moreover, the inhibitory effect of MS-PPOH on the 5-HT-induced contraction was obliterated in PAs of chronic hypoxic trpv4−/− mice. These results suggest that TRPV4 contributes to the enhanced 5-HT-induced vasoconstriction in chronic hypoxic PAs, in part via the CYP-EET-TRPV4 pathway. Our results further support the notion that manipulation of TRPV4 function may offer a novel therapeutic strategy for the treatment of hypoxia-related pulmonary hypertension.
Collapse
|
81
|
Scheraga RG, Abraham S, Grove LM, Southern BD, Crish JF, Perelas A, McDonald C, Asosingh K, Hasday JD, Olman MA. TRPV4 Protects the Lung from Bacterial Pneumonia via MAPK Molecular Pathway Switching. THE JOURNAL OF IMMUNOLOGY 2020; 204:1310-1321. [PMID: 31969384 DOI: 10.4049/jimmunol.1901033] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/22/2019] [Indexed: 12/11/2022]
Abstract
Mechanical cell-matrix interactions can drive the innate immune responses to infection; however, the molecular underpinnings of these responses remain elusive. This study was undertaken to understand the molecular mechanism by which the mechanosensitive cation channel, transient receptor potential vanilloid 4 (TRPV4), alters the in vivo response to lung infection. For the first time, to our knowledge, we show that TRPV4 protects the lung from injury upon intratracheal Pseudomonas aeruginosa in mice. TRPV4 functions to enhance macrophage bacterial clearance and downregulate proinflammatory cytokine secretion. TRPV4 mediates these effects through a novel mechanism of molecular switching of LPS signaling from predominant activation of the MAPK, JNK, to that of p38. This is accomplished through the activation of the master regulator of inflammation, dual-specificity phosphatase 1. Further, TRPV4's modulation of the LPS signal is mechanosensitive in that both upstream activation of p38 and its downstream biological consequences depend on pathophysiological range extracellular matrix stiffness. We further show the importance of TRPV4 on LPS-induced activation of macrophages from healthy human controls. These data are the first, to our knowledge, to demonstrate new roles for macrophage TRPV4 in regulating innate immunity in a mechanosensitive manner through the modulation of dual-specificity phosphatase 1 expression to mediate MAPK activation switching.
Collapse
Affiliation(s)
- Rachel G Scheraga
- Respiratory Institute, Cleveland Clinic, Cleveland, OH 44195; .,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Susamma Abraham
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Lisa M Grove
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Brian D Southern
- Respiratory Institute, Cleveland Clinic, Cleveland, OH 44195.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - James F Crish
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | | | - Christine McDonald
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Kewal Asosingh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Jeffrey D Hasday
- Department of Pulmonary and Critical Care, University of Maryland, Baltimore, MD 21201
| | - Mitchell A Olman
- Respiratory Institute, Cleveland Clinic, Cleveland, OH 44195; .,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| |
Collapse
|
82
|
Accelerated osteoblastic differentiation in patient-derived dental pulp stem cells carrying a gain-of-function mutation of TRPV4 associated with metatropic dysplasia. Biochem Biophys Res Commun 2020; 523:841-846. [PMID: 31954514 DOI: 10.1016/j.bbrc.2019.12.123] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 12/30/2019] [Indexed: 11/20/2022]
Abstract
Metatropic dysplasia (MD) is a congenital skeletal dysplasia characterized by severe platyspondyly and dumbbell-like long-bone deformities. These skeletal phenotypes are predominantly caused by autosomal dominant gain-of-function (GOF) mutations in transient receptor potential vanilloid 4 (TRPV4), which encodes a nonselective Ca2+-permeable cation channel. Previous studies have shown that constitutive TRPV4 channel activation leads to irregular chondrogenic proliferation and differentiation, and thus to the disorganized endochondral ossification seen in MD. Therefore, the present study investigated the role of TRPV4 in osteoblast differentiation and MD pathogenesis. Specifically, the behavior of osteoblasts differentiated from patient-derived dental pulp stem cells carrying a heterozygous single base TRPV4 mutation, c.1855C > T (p.L619F) was compared to that of osteoblasts differentiated from isogenic control cells (in which the mutation was corrected using the CRISPR/Cas9 system). The mutant osteoblasts exhibited enhanced calcification (indicated by intense Alizarin Red S staining), increased intracellular Ca2+ levels, strongly upregulated runt-related transcription factor 2 and osteocalcin expression, and increased expression and nuclear translocation of nuclear factor-activated T cell c1 (NFATc1) compared to control cells. These results suggest that the analyzed TRPV4 GOF mutation disrupts osteoblastic differentiation and induces MD-associated disorganized endochondral ossification by increasing Ca2+/NFATc1 pathway activity. Thus, inhibiting the NFATc1 pathway may be a promising potential therapeutic strategy to attenuate skeletal deformities in MD.
Collapse
|
83
|
Duan J, Xie J, Deng T, Xie X, Liu H, Li B, Chen M. Exposure to both formaldehyde and high relative humidity exacerbates allergic asthma by activating the TRPV4-p38 MAPK pathway in Balb/c mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 256:113375. [PMID: 31662264 DOI: 10.1016/j.envpol.2019.113375] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 09/20/2019] [Accepted: 10/09/2019] [Indexed: 06/10/2023]
Abstract
Some studies have indicated that formaldehyde, a ubiquitous environmental pollutant, can induce or aggravate allergic asthma. Epidemiological studies have also shown that the relative humidity indoors may be an independent and a key factor associated with the aggravation of allergic asthma. However, the synergy of humidity and formaldehyde on allergic asthma and the mechanism underlying this effect remain largely unknown. In this study, we aim to determine the effect of high relative humidity and/or formaldehyde exposure on allergic asthma and explore the underlying mechanisms. Male Balb/c mice were modeled with ovalbumin (OVA) and exposure to 0.5 mg/m3 formaldehyde and/or different relative humidity (60%/75%/90%). Histopathological changes, pulmonary function, Th1/Th2 balance, the status of mucus hypersecretion and the levels of inflammatory factors were detected to assess the exacerbation of allergic asthma. The levels of the transient receptor potential vanilloid 4 (TRPV4), calcium ion and the activation of p38 mitogen-activated protein kinases (p38 MAPK) were detected to explore the underlying mechanisms. The results showed that exposure to high relative humidity or to 0.5 mg/m3 formaldehyde alone had a slight, but not significant, affect on allergic asthma. However, the pathological response and airway hyperresponsiveness (AHR) were greatly aggravated by simultaneous exposure to 0.5 mg/m3 formaldehyde and 90% relative humidity. Blocking TRPV4or p38 MAPK using HC-067047 and SB203580 respectively, effectively alleviated the exacerbation of allergic asthma induced by this simultaneous exposure to formaldehyde and high relative humidity. The results show that when formaldehyde and high relative humidity are present this can enhance the activation of the TRPV4 ion channel in the lung leading to the aggravation of the p38 MAPK activation, resulting in the exacerbation of inflammation and hypersecretion of mucus in the airways.
Collapse
Affiliation(s)
- Jiufei Duan
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Jing Xie
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Ting Deng
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Xiaoman Xie
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China
| | - Hong Liu
- Joint International Research Laboratory of Green Buildings and Built Environments (Ministry of Education), Chongqing University, Chongqing 400045, China
| | - Baizhan Li
- Joint International Research Laboratory of Green Buildings and Built Environments (Ministry of Education), Chongqing University, Chongqing 400045, China
| | - Mingqing Chen
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, Hubei, China.
| |
Collapse
|
84
|
Girard BM, Campbell SE, Perkins M, Hsiang H, Tooke K, Drescher C, Hennig GW, Heppner TJ, Nelson MT, Vizzard MA. TRPV4 blockade reduces voiding frequency, ATP release, and pelvic sensitivity in mice with chronic urothelial overexpression of NGF. Am J Physiol Renal Physiol 2019; 317:F1695-F1706. [PMID: 31630542 DOI: 10.1152/ajprenal.00147.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Transient receptor potential vanilloid family member 4 (TRPV4) transcript and protein expression increased in the urinary bladder and lumbosacral dorsal root ganglia of transgenic mice with chronic urothelial overexpression of nerve growth factor (NGF-OE). We evaluated the functional role of TRPV4 in bladder function with open-outlet cystometry, void spot assays, and natural voiding (Urovoid) assays with the TRPV4 antagonist HC-067047 (1 μM) or vehicle in NGF-OE and littermate wild-type (WT) mice. Blockade of TRPV4 at the level of the urinary bladder significantly (P ≤ 0.01) increased the intercontraction interval (2.2-fold) and void volume (2.6-fold) and decreased nonvoiding contractions (3.0-fold) in NGF-OE mice, with lesser effects (1.3-fold increase in the intercontraction interval and 1.3-fold increase in the void volume) in WT mice. Similar effects of TRPV4 blockade on bladder function in NGF-OE mice were demonstrated with natural voiding assays. Intravesical administration of HC-067047 (1 µM) significantly (P ≤ 0.01) reduced pelvic sensitivity in NGF-OE mice but was without effect in littermate WT mice. Blockade of urinary bladder TRPV4 or intravesical infusion of brefeldin A significantly (P ≤ 0.01) reduced (2-fold) luminal ATP release from the urinary bladder in NGF-OE and littermate WT mice. The results of the present study suggest that TRPV4 contributes to luminal ATP release from the urinary bladder and increased voiding frequency and pelvic sensitivity in NGF-OE mice.
Collapse
Affiliation(s)
- Beatrice M Girard
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Susan E Campbell
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Megan Perkins
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Harrison Hsiang
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Katharine Tooke
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Carolyn Drescher
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Grant W Hennig
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Thomas J Heppner
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Mark T Nelson
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Margaret A Vizzard
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
85
|
Kitsuki T, Yoshimoto RU, Aijima R, Hatakeyama J, Cao AL, Zhang JQ, Ohsaki Y, Mori Y, Kido MA. Enhanced junctional epithelial permeability in TRPV4-deficient mice. J Periodontal Res 2019; 55:51-60. [PMID: 31343743 PMCID: PMC7027751 DOI: 10.1111/jre.12685] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/25/2019] [Accepted: 06/29/2019] [Indexed: 01/01/2023]
Abstract
Background and Objective As the interface between the oral cavity and the teeth, the junctional epithelial barrier is critical for gingival defense. The junctional epithelium is subject to mechanical stresses from biting force or external insults such as bacterial attacks, but little is known about the effects of mechanical stimuli on epithelial functions. Transient receptor potential vanilloid 4 (TRPV4) functions as a mechanosensitive nonselective cation channel. In the present study, based on marked expression of TRPV4 in the mouse junctional epithelium, we aimed to clarify the putative links between TRPV4 and junctional complexes in the junctional epithelium. Methods and Results Histological observations revealed that the junctional epithelium in TRPV4‐deficient (TRPV4−/−) mice had wider intercellular spaces than that in wild‐type (TRPV4+/+) mice. Exogenous tracer penetration in the junctional epithelium was greater in TRPV4−/− mice than in TRPV4+/+ mice, and immunoreactivity for adherens junction proteins was suppressed in TRPV4−/− mice compared with TRPV4+/+ mice. Analysis of a mouse periodontitis model showed greater bone volume loss in TRPV4−/− mice compared with TRPV4+/+ mice, indicating that an epithelial barrier deficiency in TRPV4−/− mice may be associated with periodontal complications. Conclusion The present findings identify a crucial role for TRPV4 in the formation of adherens junctions in the junctional epithelium, which could regulate its permeability. TRPV4 may be a candidate pharmacological target to combat periodontal diseases.
Collapse
Affiliation(s)
- Tomoko Kitsuki
- Molecular Cell Biology and Oral Anatomy, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan.,Oral and Maxillofacial Surgery, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Reiko U Yoshimoto
- Division of Histology and Neuroanatomy, Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Reona Aijima
- Division of Histology and Neuroanatomy, Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Junko Hatakeyama
- Molecular Cell Biology and Oral Anatomy, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan.,Department of Operative Dentistry and Endodontology, Fukuoka Dental College, Fukuoka, Japan
| | - Ai-Lin Cao
- Division of Histology and Neuroanatomy, Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Jing-Qi Zhang
- Molecular Cell Biology and Oral Anatomy, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yasuyoshi Ohsaki
- Division of Histology and Neuroanatomy, Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Yoshihide Mori
- Oral and Maxillofacial Surgery, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Mizuho A Kido
- Division of Histology and Neuroanatomy, Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
86
|
McDonough RC, Shoga JS, Price C. DREADD-based synthetic control of chondrocyte calcium signaling in vitro. J Orthop Res 2019; 37:1518-1529. [PMID: 30908734 DOI: 10.1002/jor.24285] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 03/08/2019] [Indexed: 02/04/2023]
Abstract
Calcium is a critical second messenger involved in chondrocyte mechanotransduction. Several distinct calcium signaling mechanisms implicated in chondrocyte mechanotransduction have been identified using mechanical perturbations or soluble signaling factors. However, these commonly used stimuli can lack specificity in the mechanisms by which they initiate calcium signaling. Synthetic tools allowing for more precise and selective regulation of calcium signaling, such as the engineered G-protein-coupled receptors known as DREADDs (Designer Receptors Exclusively Activated by Designer Drugs), may better assist in isolating the roles of intracellular calcium ([Ca2+ ]i ) and cell activation in chondrocyte biology. One DREADD, hM3Dq, is solely activated by clozapine N-oxide (CNO) and regulates calcium activation through the Gq -PLCβ-IP3 -ER pathway. Here, hM3Dq-transfected ATDC5 cells were treated with CNO (100 nM-1 μM) to establish the feasibility of using Gq -DREADDs to drive [Ca2+ ]i activation in chondrocyte-like cells. CNO administration resulted in a coordinated, dose-dependent, and transient calcium response in hM3Dq-transfected cells that resulted primarily from calcium release from the ER. Following activation via CNO administration, hM3Dq-ATDC5 cells exhibited refractory behavior and required a 4-h wash-out period to recover hM3Dq-mediated signaling. However, hM3Dq inactivation did not inhibit alternative calcium activation mechanisms in ATDC5 cells (via GSK101 or hypo-osmotic shock), nor did CNO-driven calcium signaling negatively impact ATDC5 cell health. This study established the successful use of hM3Dq for the safe, targeted, and well-controlled activation of calcium signaling in ATDC5 cells and its use as a potential tool for assessing clinically significant questions regarding calcium signaling in chondrocyte biology, cartilage pathology, and cartilage tissue engineering. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1518-1529, 2019.
Collapse
Affiliation(s)
- Ryan C McDonough
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, 19716, DE
| | - Janty S Shoga
- Department of Biomechanics and Movement Science, University of Delaware, Newark, DE
| | - Christopher Price
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, 19716, DE.,Department of Biomechanics and Movement Science, University of Delaware, Newark, DE
| |
Collapse
|
87
|
Clinical Pharmacokinetics, Safety, and Tolerability of a Novel, First-in-Class TRPV4 Ion Channel Inhibitor, GSK2798745, in Healthy and Heart Failure Subjects. Am J Cardiovasc Drugs 2019; 19:335-342. [PMID: 30637626 DOI: 10.1007/s40256-018-00320-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION AND OBJECTIVE Pulmonary capillary endothelial transient receptor potential vanilloid 4 (TRPV4) channel plays a critical role in mediating the development of cardiogenic pulmonary edema. GSK2798745 is a first-in-class, highly potent, selective, orally active TRPV4 channel blocker being evaluated in a first-time-in-humans study (NCT02119260). METHODS GSK2798745 was administered in a randomized, placebo-controlled study design to healthy volunteers in three separate cohorts as single escalating doses, with and without food, and as once-daily repeat doses for up to 14 days, respectively. Two cohorts of subjects with mild to moderate heart failure were also administered GSK2798745 once daily for up to 7 days. Safety, tolerability, and systemic exposure data were collected. RESULTS No significant safety issues or serious adverse events were observed with GSK2798745 in healthy volunteers and patients with heart failure. GSK2798745 systemic exposure data demonstrated linear pharmacokinetics up to 12.5 mg, less than twofold accumulation with once-daily dosing, and a systemic half-life of ~ 13 h. There was a slight increase in GSK2798745 exposure [14% increase in area under the plasma concentration-time curve (AUC) and 9% increase in maximum observed plasma concentration (Cmax)] after administration with a high-fat meal. CONCLUSIONS GSK2798745 was well-tolerated in healthy volunteers and patients with stable heart failure. The safety and exposure data obtained in this study allow further evaluation of the drug in long-term clinical studies in heart failure as well as other indications.
Collapse
|
88
|
Andersson KE. TRP Channels as Lower Urinary Tract Sensory Targets. Med Sci (Basel) 2019; 7:E67. [PMID: 31121962 PMCID: PMC6572419 DOI: 10.3390/medsci7050067] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/19/2019] [Accepted: 05/20/2019] [Indexed: 12/13/2022] Open
Abstract
Several members of the transient receptor potential (TRP) superfamily, including TRPV1, TRPV2, TRPV4, TRM4, TRPM8 and TRPA1, are expressed in the lower urinary tract (LUT), not only in neuronal fibers innervating the bladder and urethra, but also in the urothelial and muscular layers of the bladder and urethral walls. In the LUT, TRP channels are mainly involved in nociception and mechanosensory transduction. Animal studies have suggested the therapeutic potential of several TRP channels for the treatment of both bladder over- and underactivity and bladder pain disorders,; however translation of this finding to clinical application has been slow and the involvement of these channels in normal human bladder function, and in various pathologic states have not been established. The development of selective TRP channel agonists and antagonists is ongoing and the use of such agents can be expected to offer new and important information concerning both normal physiological functions and possible therapeutic applications.
Collapse
Affiliation(s)
- Karl-Erik Andersson
- Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston Salem, NC 27101, USA.
- Institute of Laboratory Medicine, Lund University, 223 62 Lund, Sweden.
| |
Collapse
|
89
|
Goutsou S, Tsakona C, Polia A, Moutafidi A, Zolota V, Gatzounis G, Assimakopoulou M. Transient receptor potential vanilloid (TRPV) channel expression in meningiomas: prognostic and predictive significance. Virchows Arch 2019; 475:105-114. [DOI: 10.1007/s00428-019-02584-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/24/2019] [Accepted: 05/07/2019] [Indexed: 01/03/2023]
|
90
|
Goswami R, Arya RK, Biswas D, Zhu X, Rahaman SO. Transient Receptor Potential Vanilloid 4 Is Required for Foreign Body Response and Giant Cell Formation. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1505-1512. [PMID: 31121133 DOI: 10.1016/j.ajpath.2019.04.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/05/2019] [Accepted: 04/11/2019] [Indexed: 12/11/2022]
Abstract
The presence of biomaterials and devices implanted into soft tissue is associated with development of a foreign body response (FBR), a chronic inflammatory condition that can ultimately lead to implant failure, which may cause harm to or death of the patient. Development of FBR includes activation of macrophages at the tissue-implant interface, generation of destructive foreign body giant cells (FBGCs), and generation of fibrous tissue that encapsulates the implant. However, the mechanisms underlying the FBR remain poorly understood, as neither the materials composing the implants nor their chemical properties can explain triggering of the FBR. Herein, we report that genetic ablation of transient receptor potential vanilloid 4 (TRPV4), a Ca2+-permeable mechanosensitive cation channel in the transient receptor potential vanilloid family, protects TRPV4 knockout mice from FBR-related events. The mice showed diminished collagen deposition along with reduced macrophage accumulation and FBGC formation compared with wild-type mice in a s.c. implantation model. Analysis of macrophage markers in spleen tissues and peritoneal cavity showed that the TRPV4 deficiency did not impair basal macrophage maturation. Furthermore, genetic deficiency or pharmacologic antagonism of TRPV4 blocked cytokine-induced FBGC formation, which was restored by lentivirus-mediated TRPV4 reintroduction. Taken together, these results suggest an important, previously unknown, role for TRPV4 in FBR.
Collapse
Affiliation(s)
- Rishov Goswami
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| | - Rakesh K Arya
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| | - Debabrata Biswas
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Xiaoping Zhu
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland
| | - Shaik O Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland.
| |
Collapse
|
91
|
Nonaka K, Han X, Kato H, Sato H, Yamaza H, Hirofuji Y, Masuda K. Novel gain-of-function mutation of TRPV4 associated with accelerated chondrogenic differentiation of dental pulp stem cells derived from a patient with metatropic dysplasia. Biochem Biophys Rep 2019; 19:100648. [PMID: 31463371 PMCID: PMC6709385 DOI: 10.1016/j.bbrep.2019.100648] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 04/22/2019] [Accepted: 05/03/2019] [Indexed: 01/16/2023] Open
Abstract
Metatropic dysplasia is a congenital skeletal dysplasia characterized by severe platyspondyly, dumbbell-like deformity of long tubular bones, and progressive kyphoscoliosis with growth. It is caused by mutations in the gene TRPV4, encoding the transient receptor potential vanilloid 4, which acts as a calcium channel. Many heterozygous single base mutations of this gene have been associated with the disorder, showing autosomal dominant inheritance. Although abnormal endochondral ossification has been observed by histological examination of bone in a patient with lethal metatropic dysplasia, the etiology of the disorder remains largely unresolved. As dental pulp stem cells (DPSCs) are mesenchymal stem cells that differentiate into bone lineage cells, DPSCs derived from patients with congenital skeletal dysplasia might be useful as a disease-specific cellular model for etiological investigation. The purpose of this study was to clarify the pathological association between TRPV4 mutation and chondrocyte differentiation by analyzing DPSCs from a patient with non-lethal metatropic dysplasia. We identified a novel heterozygous single base mutation, c.1855C>T in TRPV4. This was predicted to be a missense mutation, p.L619F, in putative transmembrane segment 5. The mutation was repaired by CRISPR/Cas9 system to obtain isogenic control DPSCs for further analysis. The expression of stem cell markers and fibroblast-like morphology were comparable between patient-derived mutant and control DPSCs, although expression of TRPV4 was lower in mutant DPSCs than control DPSCs. Despite the lower TRPV4 expression in mutant DPSCs, the intracellular Ca2+ level was comparable at the basal level between mutant and control DPSCs, while its level was markedly higher following stimulation with 4α-phorbol 12,13-didecanoate (4αPDD), a specific agonist for TRPV4, in mutant DPSCs than in control DPSCs. In the presence of 4αPDD, we observed accelerated early chondrocyte differentiation and upregulated mRNA expression of SRY-box 9 (SOX9) in mutant DPSCs. Our findings suggested that the novel missense mutation c.1855C>T of TRPV4 was a gain-of-function mutation leading to enhanced intracellular Ca2+ level, which was associated with accelerated chondrocyte differentiation and SOX9 upregulation. Our results also suggest that patient-derived DPSCs can be a useful disease-specific cellular model for elucidating the pathological mechanism of metatropic dysplasia. Dental pulp stem cells from a patient with metatropic dysplasia were analyzed. A novel heterozygous single base mutation, c.1855C > T, was identified in TRPV4. The mutation was repaired by gene editing to obtain isogenic control cells. The intracellular Ca2+ level was enhanced in mutant cells by 4αPDD. Chondrocyte differentiation was enhanced in mutant cells by 4αPDD.
Collapse
Affiliation(s)
- Kentaro Nonaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Xu Han
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Hiroki Kato
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Hiroshi Sato
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Haruyoshi Yamaza
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yuta Hirofuji
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Keiji Masuda
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Maidashi 3-1-1, Higashi-Ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
92
|
Tomilin V, Mamenko M, Zaika O, Wingo CS, Pochynyuk O. TRPV4 deletion protects against hypokalemia during systemic K + deficiency. Am J Physiol Renal Physiol 2019; 316:F948-F956. [PMID: 30838874 PMCID: PMC6580258 DOI: 10.1152/ajprenal.00043.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/05/2019] [Accepted: 03/05/2019] [Indexed: 12/18/2022] Open
Abstract
Tight regulation of K+ balance is fundamental for normal physiology. Reduced dietary K+ intake, which is common in Western diets, often leads to hypokalemia and associated cardiovascular- and kidney-related pathologies. The distal nephron, and, specifically, the collecting duct (CD), is the major site of controlled K+ reabsorption via H+-K+-ATPase in the state of dietary K+ deficiency. We (Mamenko MV, Boukelmoune N, Tomilin VN, Zaika OL, Jensen VB, O'Neil RG, Pochynyuk OM. Kidney Int 91: 1398-1409, 2017) have previously demonstrated that the transient receptor potential vanilloid type 4 (TRPV4) Ca2+ channel, abundantly expressed in the CD, contributes to renal K+ handling by promoting flow-induced K+ secretion. Here, we investigated a potential role of TRPV4 in controlling H+-K+-ATPase-dependent K+ reabsorption in the CD. Treatment with a K+-deficient diet (<0.01% K+) for 7 days reduced serum K+ levels in wild-type (WT) mice from 4.3 ± 0.2 to 3.3 ± 0.2 mM but not in TRPV4-/- mice (4.3 ± 0.1 and 4.2 ± 0.3 mM, respectively). Furthermore, we detected a significant reduction in 24-h urinary K+ levels in TRPV4-/- compared with WT mice upon switching to K+-deficient diet. TRPV4-/- animals also had significantly more acidic urine on a low-K+ diet, but not on a regular (0.9% K+) or high-K+ (5% K+) diet, which is consistent with increased H+-K+-ATPase activity. Moreover, we detected a greatly accelerated H+-K+-ATPase-dependent intracellular pH extrusion in freshly isolated CDs from TRPV4-/- compared with WT mice fed a K+-deficient diet. Overall, our results demonstrate a novel kaliuretic role of TRPV4 by inhibiting H+-K+-ATPase-dependent K+ reabsorption in the CD. We propose that TRPV4 inhibition could be a novel strategy to manage certain hypokalemic states in clinical settings.
Collapse
Affiliation(s)
- Viktor Tomilin
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston , Houston, Texas
| | - Mykola Mamenko
- Department of Physiology, Medical College of Georgia, Augusta University , Augusta, Georgia
| | - Oleg Zaika
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston , Houston, Texas
| | - Charles S Wingo
- Division of Nephrology, Hypertension and Transplantation, Department of Medicine, University of Florida , Gainesville, Florida
- North Florida/South Georgia Veterans Health System, Gainesville, Florida
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston , Houston, Texas
| |
Collapse
|
93
|
Rahman M, Sun R, Mukherjee S, Nilius B, Janssen LJ. TRPV4 Stimulation Releases ATP via Pannexin Channels in Human Pulmonary Fibroblasts. Am J Respir Cell Mol Biol 2019; 59:87-95. [PMID: 29393654 DOI: 10.1165/rcmb.2017-0413oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
We previously described several ionic conductances in human pulmonary fibroblasts, including one activated by two structurally distinct TRPV4 (transient receptor potential, vanilloid-type, subtype 4)-channel agonists: 4αPDD (4α-phorbol-12,13-didecanoate) and GSK1016790A. However, the TRPV4-activated current exhibited peculiar properties: it developed slowly over many minutes, exhibited reversal potentials that could vary by tens of millivolts even within a given cell, and was not easily reversed by subsequent addition of two distinct TRPV4-selective blockers (RN-1734 and HC-067047). In this study, we characterized that conductance more carefully. We found that 4αPDD stimulated a delayed release of ATP into the extracellular space, which was reduced by genetic silencing of pannexin expression, and that the 4αPDD-evoked current could be blocked by apyrase (which rapidly degrades ATP) or by the P2Y purinergic receptor/channel blocker pyridoxalphosphate-6-azophenyl-2',4'-disulphonic acid (PPADS), and could be mimicked by exogenous addition of ATP. In addition, we found that the 4αPDD-evoked current was blocked by pretreatment with RN-1734 or HC-067047, by Gd3+ or La3+, or by two distinct blockers of pannexin channels (carbenoxolone and probenecid), but not by a blocker of connexin hemichannels (flufenamic acid). We also found expression of TRPV4- and pannexin-channel proteins. 4αPDD markedly increased calcium flashing in our cells. The latter was abrogated by the P2Y channel blocker PPADS, and the 4αPDD-evoked current was eliminated by loading the cytosol with 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid or by inhibiting Ca2+/calmodulin-sensitive kinase II using KN93. Altogether, we interpret these findings as suggesting that 4αPDD triggers the release of ATP via pannexin channels, which in turn acts in an autocrine and/or paracrine fashion to stimulate PPADS-sensitive purinergic receptors on human pulmonary fibroblasts.
Collapse
Affiliation(s)
- Mozibur Rahman
- 1 Firestone Institute for Respiratory Health, St. Joseph's Hospital, and.,2 Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Rui Sun
- 1 Firestone Institute for Respiratory Health, St. Joseph's Hospital, and.,2 Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Subhendu Mukherjee
- 1 Firestone Institute for Respiratory Health, St. Joseph's Hospital, and.,2 Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Bernd Nilius
- 3 Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Luke J Janssen
- 1 Firestone Institute for Respiratory Health, St. Joseph's Hospital, and.,2 Department of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| |
Collapse
|
94
|
Distasi C, Dionisi M, Ruffinatti FA, Gilardino A, Bardini R, Antoniotti S, Catalano F, Bassino E, Munaron L, Martra G, Lovisolo D. The interaction of SiO 2 nanoparticles with the neuronal cell membrane: activation of ionic channels and calcium influx. Nanomedicine (Lond) 2019; 14:575-594. [PMID: 30810075 DOI: 10.2217/nnm-2018-0256] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIM To clarify the mechanisms of interaction between SiO2 nanoparticles (NPs) and the plasma membrane of GT1-7 neuroendocrine cells, with focus on the activation of calcium-permeable channels, responsible for the long lasting calcium influx and modulation of the electrical activity in these cells. MATERIALS & METHODS Nontoxic doses of SiO2 NPs were administered to the cells. Calcium imaging and patch clamp techniques were combined with a pharmacological approach. RESULTS TRPV4, Cx and Panx-like channels are the major components of the NP-induced inward currents. Preincubation with the antioxidant N-acetyl-L-cysteine strongly reduced the [Ca2+]i increase. CONCLUSION These findings suggest that SiO2 NPs directly activate a complex set of calcium-permeable channels, possibly by catalyzing free radical production.
Collapse
Affiliation(s)
- Carla Distasi
- Department of Pharmaceutical Sciences, University of Piemonte Orientale 'A. Avogadro', Via Bovio 6, 28100 Novara, Italy
| | - Marianna Dionisi
- Department of Pharmaceutical Sciences, University of Piemonte Orientale 'A. Avogadro', Via Bovio 6, 28100 Novara, Italy
| | | | - Alessandra Gilardino
- Department of Life Sciences & Systems Biology, University of Torino, via Accademia Albertina 23, 10123 Torino, Italy
| | - Roberta Bardini
- Department of Life Sciences & Systems Biology, University of Torino, via Accademia Albertina 23, 10123 Torino, Italy.,Department of Control & Computer Engineering, Polytechnic University of Turin, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Susanna Antoniotti
- Department of Life Sciences & Systems Biology, University of Torino, via Accademia Albertina 23, 10123 Torino, Italy
| | - Federico Catalano
- Department of Chemistry, Torino, University of Torino, Via P. Giuria 9, 10125, Italy.,Italian Institute of Technology, Central Research Laboratories, Via Morego 30, 16163 Genova, Italy
| | - Eleonora Bassino
- Department of Life Sciences & Systems Biology, University of Torino, via Accademia Albertina 23, 10123 Torino, Italy
| | - Luca Munaron
- Department of Life Sciences & Systems Biology, University of Torino, via Accademia Albertina 23, 10123 Torino, Italy
| | - Gianmario Martra
- Department of Control & Computer Engineering, Polytechnic University of Turin, Corso Duca degli Abruzzi 24, 10129 Torino, Italy.,NIS Interdepartmental Center, University of Torino, Italy
| | - Davide Lovisolo
- Department of Life Sciences & Systems Biology, University of Torino, via Accademia Albertina 23, 10123 Torino, Italy
| |
Collapse
|
95
|
Huang R, Wang F, Yang Y, Ma W, Lin Z, Cheng N, Long Y, Deng S, Li Z. Recurrent activations of transient receptor potential vanilloid-1 and vanilloid-4 promote cellular proliferation and migration in esophageal squamous cell carcinoma cells. FEBS Open Bio 2019; 9:206-225. [PMID: 30761248 PMCID: PMC6356177 DOI: 10.1002/2211-5463.12570] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/19/2018] [Accepted: 10/23/2018] [Indexed: 12/27/2022] Open
Abstract
Some members of the transient receptor potential vanilloid (TRPV) subfamily of cation channels are thermosensitive. Earlier studies have revealed the distribution and functions of these thermo‐TRPVs (TRPV1–4) in various organs, but their expression and function in the human esophagus are not fully understood. Here, we probed for the expression of the thermo‐TRPVs in one nontumor human esophageal squamous cell line and two esophageal squamous cell carcinoma (ESCC) cell lines. TRPV1, TRPV2, and TRPV4 proteins were found to be upregulated in ESCC cells, while TRPV3 was not detectable in any of these cell lines. Subsequently, channel function was evaluated via monitoring of Ca2+ transients by Ca2+ imaging and nonselective cation channel currents were recorded by whole‐cell patch clamp. We found that TRPV4 was activated by heat at 28 °C–35 °C, whereas TRPV1 and TRPV2 were activated by higher, noxious temperatures (44 °C and 53 °C, respectively). Furthermore, TRPV1 was activated by capsaicin (EC50 = 20.32 μm), and this effect was antagonized by AMG9810; TRPV2 was activated by a newly developed cannabinoid compound, O1821, and inhibited by tranilast. In addition, TRPV4 was activated by hypotonic solutions (220 m Osm), and this effect was abolished by ruthenium red. The effects of TRPV1 and TRPV4 on ESCC were also explored. Our data, for the first time, showed that the overactivation of TRPV1 and TRPV4 promoted the proliferation and/or migration of ESCC cells. In summary, TRPV1, TRPV2, and TRPV4 were functionally expressed in human esophageal squamous cells, and thermo‐TRPVs might play an important role in the development of ESCC.
Collapse
Affiliation(s)
- Rongqi Huang
- Key Laboratory of Regenerative Biology Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine Guangzhou Institutes of Biomedicine and Health Chinese Academy of Sciences Guangzhou China.,University of Chinese Academy of Sciences Beijing China
| | - Fei Wang
- Key Laboratory of Regenerative Biology Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine Guangzhou Institutes of Biomedicine and Health Chinese Academy of Sciences Guangzhou China
| | - Yuchen Yang
- Key Laboratory of Regenerative Biology Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine Guangzhou Institutes of Biomedicine and Health Chinese Academy of Sciences Guangzhou China
| | - Wenbo Ma
- Key Laboratory of Regenerative Biology Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine Guangzhou Institutes of Biomedicine and Health Chinese Academy of Sciences Guangzhou China
| | - Zuoxian Lin
- Key Laboratory of Regenerative Biology Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine Guangzhou Institutes of Biomedicine and Health Chinese Academy of Sciences Guangzhou China
| | - Na Cheng
- Key Laboratory of Regenerative Biology Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine Guangzhou Institutes of Biomedicine and Health Chinese Academy of Sciences Guangzhou China.,Department of Anatomy and Neurobiology Xiangya School of Medicine Central South University Changsha China
| | - Yan Long
- Key Laboratory of Regenerative Biology Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine Guangzhou Institutes of Biomedicine and Health Chinese Academy of Sciences Guangzhou China
| | - Sihao Deng
- Department of Anatomy and Neurobiology Xiangya School of Medicine Central South University Changsha China
| | - Zhiyuan Li
- Key Laboratory of Regenerative Biology Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine Guangzhou Institutes of Biomedicine and Health Chinese Academy of Sciences Guangzhou China.,University of Chinese Academy of Sciences Beijing China.,Department of Anatomy and Neurobiology Xiangya School of Medicine Central South University Changsha China.,GZMU-GIBH Joint School of Life Sciences Guangzhou Medical University China
| |
Collapse
|
96
|
Gao P, Yan Z, Zhu Z. The role of adipose TRP channels in the pathogenesis of obesity. J Cell Physiol 2019; 234:12483-12497. [PMID: 30618095 DOI: 10.1002/jcp.28106] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 12/07/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Peng Gao
- Department of Hypertension and Endocrinology Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Zhencheng Yan
- Department of Hypertension and Endocrinology Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension Chongqing China
| |
Collapse
|
97
|
Faye E, Modaff P, Pauli R, Legare J. Combined Phenotypes of Spondylometaphyseal Dysplasia-Kozlowski Type and Charcot-Marie-Tooth Disease Type 2C Secondary to a TRPV4 Pathogenic Variant. Mol Syndromol 2018; 10:154-160. [PMID: 31191204 DOI: 10.1159/000495778] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2018] [Indexed: 11/19/2022] Open
Abstract
TRPV4, a nonselective calcium permeable ion channel, is expressed broadly in many organs including bone and neurons. Pathogenic variants in TRPV4 are known to cause both a spectrum of skeletal dysplasias and neuropathies. Recent publications have documented a few patients who have a combined phenotype of skeletal dysplasia and neuropathy secondary to TRPV4 pathogenic variants. We present an additional patient who has an overlapping neuromuscular and skeletal phenotype secondary to a TRPV4 pathogenic variant. The patient has spondylometaphyseal dysplasia-Kozlowski type and Charcot-Marie-Tooth disease type 2C. This and prior reports illustrate that TRPV4-related skeletal dysplasias and TRPV4-related neuropathies are not fully distinct disorders secondary to unique sets of pathogenic variants as originally postulated, but rather are 2 phenotypes on the same spectrum that may or may not overlap. We suggest that evaluation for patients presenting with any TRPV4-related disorder include assessment for both skeletal and neurological findings.
Collapse
Affiliation(s)
- Eden Faye
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Peggy Modaff
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Richard Pauli
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Janet Legare
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
98
|
Sharma S, Goswami R, Rahaman SO. The TRPV4-TAZ mechanotransduction signaling axis in matrix stiffness- and TGFβ1-induced epithelial-mesenchymal transition. Cell Mol Bioeng 2018; 12:139-152. [PMID: 31681446 DOI: 10.1007/s12195-018-00565-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Introduction The implantation of biomaterials into soft tissue leads to the development of foreign body response, a non-specific inflammatory condition that is characterized by the presence of fibrotic tissue. Epithelial-mesenchymal transition (EMT) is a key event in development, fibrosis, and oncogenesis. Emerging data support a role for both a mechanical signal and a biochemical signal in EMT. We hypothesized that transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive channel, is a mediator of EMT. Methods Normal human primary epidermal keratinocytes (NHEKs) were seeded on collagen-coated plastic plates or varied stiffness polyacrylamide gels in the presence or absence of TGFβ1, Immunofluorescence, immunoblot, and polymerase chain reaction analysis were performed to determine expression level of EMT markers and signaling proteins. Knock-down of TRPV4 function was achieved by siRNA transfection or by GSK2193874 treatment. Results We found that knock-down of TRPV4 blocked both matrix stiffness- and TGFβ1-induced EMT in NHEKs. In a murine skin fibrosis model, TRPV4 deletion resulted in decreased expression of the mesenchymal marker, α-SMA, and increased expression of epithelial marker, E-cadherin. Mechanistically, our data showed that: i) TRPV4 was essential for the nuclear translocation of TAZ in response to matrix stiffness and TGFβ1; ii) Antagonism of TRPV4 inhibited both matrix stiffness-induced and TGFβ1-induced expression of TAZ proteins; and iii) TRPV4 antagonism suppressed both matrix stiffness-induced and TGFβ1-induced activation of Smad2/3, but not of AKT. Conclusions These data identify a novel role for TRPV4-TAZ mechanotransduction signaling axis in regulating EMT in NHEKs in response to both matrix stiffness and TGFβ1.
Collapse
Affiliation(s)
- Shweta Sharma
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742 USA
| | - Rishov Goswami
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742 USA
| | - Shaik O Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742 USA
| |
Collapse
|
99
|
Pero JE, Matthews JM, Behm DJ, Brnardic EJ, Brooks C, Budzik BW, Costell MH, Donatelli CA, Eisennagel SH, Erhard K, Fischer MC, Holt DA, Jolivette LJ, Li H, Li P, McAtee JJ, McCleland BW, Pendrak I, Posobiec LM, Rivera KL, Rivero RA, Roethke TJ, Sender MR, Shu A, Terrell LR, Vaidya K, Xu X, Lawhorn BG. Design and Optimization of Sulfone Pyrrolidine Sulfonamide Antagonists of Transient Receptor Potential Vanilloid-4 with in Vivo Activity in a Pulmonary Edema Model. J Med Chem 2018; 61:11209-11220. [DOI: 10.1021/acs.jmedchem.8b01344] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
100
|
Sharma S, Goswami R, Zhang DX, Rahaman SO. TRPV4 regulates matrix stiffness and TGFβ1-induced epithelial-mesenchymal transition. J Cell Mol Med 2018; 23:761-774. [PMID: 30450767 PMCID: PMC6349341 DOI: 10.1111/jcmm.13972] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 09/14/2018] [Accepted: 09/20/2018] [Indexed: 12/20/2022] Open
Abstract
Substrate stiffness (or rigidity) of the extracellular matrix has important functions in numerous pathophysiological processes including fibrosis. Emerging data support a role for both a mechanical signal, for example, matrix stiffness, and a biochemical signal, for example, transforming growth factor β1 (TGFβ1), in epithelial‐mesenchymal transition (EMT), a process critically involved in fibrosis. Here, we report evidence showing that transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive channel, is the likely mediator of EMT in response to both TGFβ1 and matrix stiffness. Specifically, we found that: (a) genetic ablation or pharmacological inhibition of TRPV4 blocked matrix stiffness and TGFβ1‐induced EMT in normal mouse primary epidermal keratinocytes (NMEKs) as determined by changes in morphology, adhesion, migration and alterations of expression of EMT markers including E‐cadherin, N‐cadherin (NCAD) and α‐smooth muscle actin (α‐SMA), and (b) TRPV4 deficiency prevented matrix stiffness‐induced EMT in NMEKs over a pathophysiological range. Intriguingly, TRPV4 deletion in mice suppressed expression of mesenchymal markers, NCAD and α‐SMA, in a bleomycin‐induced murine skin fibrosis model. Mechanistically, we found that: (a) TRPV4 was essential for the nuclear translocation of YAP/TAZ (yes‐associated protein/transcriptional coactivator with PDZ‐binding motif) in response to matrix stiffness and TGFβ1, (b) TRPV4 deletion inhibited both matrix stiffness‐ and TGFβ1‐induced expression of YAP/TAZ proteins and (c) TRPV4 deletion abrogated both matrix stiffness‐ and TGFβ1‐induced activation of AKT, but not Smad2/3, suggesting a mechanism by which TRPV4 activity regulates EMT in NMEKs. Altogether, these data identify a novel role for TRPV4 in regulating EMT.
Collapse
Affiliation(s)
- Shweta Sharma
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| | - Rishov Goswami
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| | - David X Zhang
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Shaik O Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| |
Collapse
|