Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Hypertension ; 80(11): 2345-2356, 2023 11.
Article in English | MEDLINE | ID: mdl-37702061

ABSTRACT

BACKGROUND: Left ventricular hypertrophy is a bipolar response, starting as an adaptive response to the hemodynamic challenge, but over time develops maladaptive pathology partly due to microvascular rarefaction and impaired coronary angiogenesis. Despite the profound influence on cardiac function, the mechanotransduction mechanisms that regulate coronary angiogenesis, leading to heart failure, are not well known. METHODS: We subjected endothelial-specific knockout mice of mechanically activated ion channel, TRPV4 (transient receptor potential cation channel subfamily V member 4; TRPV4ECKO) to pressure overload via transverse aortic constriction and examined cardiac function, cardiomyocyte hypertrophy, cardiac fibrosis, and apoptosis. Further, we measured microvascular density and underlying TRPV4 mechanotransduction mechanisms using human microvascular endothelial cells, extracellular matrix gels of varying stiffness, unbiased RNA sequencing, small interfering RNA, Western blot, quantitative-PCR, and confocal immunofluorescence techniques. RESULTS: We demonstrate that endothelial-specific deletion of TRPV4 preserved cardiac function, cardiomyocyte structure, and reduced cardiac fibrosis compared with TRPV4lox/lox mice, 28 days post-transverse aortic constriction. Interestingly, comprehensive RNA sequencing analysis revealed an upregulation of proangiogenic factors (VEGFα [vascular endothelial growth factor α], NOS3 [nitric oxide synthase 3], and FGF2 [fibroblast growth factor 2]) with concomitant increase in microvascular density in TRPV4ECKO hearts after transverse aortic constriction compared with TRPV4lox/lox. Further, an increased expression of VEGFR2 (vascular endothelial growth factor receptor 2) and activation of the YAP (yes-associated protein) pathway were observed in TRPV4ECKO hearts. Mechanistically, we found that downregulation of TRPV4 in endothelial cells induced matrix stiffness-dependent activation of YAP and VEGFR2 via the Rho/Rho kinase/large tumor suppressor kinase pathway. CONCLUSIONS: Our results suggest that endothelial TRPV4 acts as a mechanical break for coronary angiogenesis, and uncoupling endothelial TRPV4 mechanotransduction attenuates pathological cardiac hypertrophy by enhancing coronary angiogenesis.


Subject(s)
Cardiomegaly , Mechanotransduction, Cellular , TRPV Cation Channels , Animals , Humans , Mice , Cardiomegaly/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Hypertrophy, Left Ventricular/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Vascular Endothelial Growth Factor A/metabolism
2.
FASEB J ; 37(10): e23199, 2023 10.
Article in English | MEDLINE | ID: mdl-37732601

ABSTRACT

Prostaglandin E2 (PGE2 ) has been implicated in counteracting fibroblast differentiation by TGFß1 during pulmonary fibrosis. However, the precise mechanism is not well understood. We show here that PGE2 via EP2 R and EP4 R inhibits the expression of mechanosensory molecules Lysyl Oxidase Like 2 (LOXL2), myocardin-related transcription factor A (MRTF-A), ECM proteins, plasminogen activation inhibitor 1 (PAI-1), fibronectin (FN), α-smooth muscle actin (α-SMA), and redox sensor (nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4)) required for TGFß1-mediated fibroblast differentiation. We further demonstrate that PGE2 inhibits fibrotic signaling via Yes-associated protein (YAP) but does so independently from its actions on SMAD phosphorylation and conserved cylindromatosis (CYLD; deubiquitinase) expression. Mechanistically, PGE2 phosphorylates/inactivates YAP downstream of EP2 R/Gαs and restrains its translocation to the nucleus, thus inhibiting its interaction with TEA domain family members (TEADs) and transcription of fibrotic genes. Importantly, pharmacological or siRNA-mediated inhibition of YAP significantly downregulates TGFß1-mediated fibrotic gene expression and myofibroblast formation. Notably, YAP expression is upregulated in the lungs of D. farinae-treated wild type (WT) mice relative to saline-treated WT mice. Our results unravel a unique role for PGE2 -YAP interactions in fibroblast differentiation, and that PGE2 /YAP inhibition can be used as a novel therapeutic target in the treatment of pathological conditions associated with myofibroblasts like asthma.


Subject(s)
Dinoprostone , YAP-Signaling Proteins , Animals , Mice , Fibroblasts , Signal Transduction , Myofibroblasts
3.
J Cell Mol Med ; 27(19): 2983-2994, 2023 10.
Article in English | MEDLINE | ID: mdl-37603611

ABSTRACT

Short-chain fatty acid butyrate is produced from the bacterial fermentation of indigestible fiber in the intestinal lumen, and it has been shown to attenuate lung inflammation in murine asthma models. Mast cells (MCs) are initiators of inflammatory response to allergens, and they play an important role in asthma. MC survival and proliferation is regulated by its growth factor stem cell factor (SCF), which acts through the receptor, KIT. It has previously been shown that butyrate attenuates the activation of MCs by allergen stimulation. However, how butyrate mechanistically influences SCF signalling to impact MC function remains unknown. Here, we report that butyrate treatment triggered the modification of MC histones via butyrylation and acetylation, and inhibition of histone deacetylase (HDAC) activity. Further, butyrate treatment caused downregulation of SCF receptor KIT and associated phosphorylation, leading to significant attenuation of SCF-mediated MC proliferation, and pro-inflammatory cytokine secretion. Mechanistically, butyrate inhibited MC function by suppressing KIT and downstream p38 and Erk phosphorylation, and it mediated these effects via modification of histones, acting as an HDAC inhibitor and not via its traditional GPR41 (FFAR3) or GPR43 (FFAR2) butyrate receptors. In agreement, the pharmacological inhibition of Class I HDAC (HDAC1/3) mirrored butyrate's effects, suggesting that butyrate impacts MC function by HDAC1/3 inhibition. Taken together, butyrate epigenetically modifies histones and downregulates the SCF/KIT/p38/Erk signalling axis, leading to the attenuation of MC function, validating its ability to suppress MC-mediated inflammation. Therefore, butyrate supplementations could offer a potential treatment strategy for allergy and asthma via epigenetic alterations in MCs.


Subject(s)
Asthma , Histones , Humans , Mice , Animals , Histones/metabolism , Mast Cells/metabolism , Butyrates/pharmacology , Histone Code , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Epigenesis, Genetic , Asthma/metabolism
5.
Article in English | MEDLINE | ID: mdl-38333200

ABSTRACT

Angiotensin II (Ang II) is a potent vasoconstrictor of vascular smooth muscle cells (VSMC) and is implicated in hypertension, but it's role in the regulation of endothelial function is not well known. We and others have previously shown that mechanically activated ion channel, Transient Receptor Potential Vanilloid 4 (TRPV4) mediates flow- and/or receptor-dependent vasodilation via nitric oxide (NO) production in endothelial cells. Ang II was demonstrated to crosstalk with TRPV4 via angiotensin 1 receptor (AT1R) and ß-arrestin signaling in epithelial and immortalized cells, however, the role of this crosstalk in endothelial cell function is not fully explored. Ang II treatment significantly downregulated TRPV4 protein expression and TRPV4-mediated Ca2+ influx in human EC without altering TRPV4 mRNA levels. Further, TRPV4-induced eNOS phosphorylation and NO production were significantly reduced in Ang II-treated human EC. Importantly, Ang II infusion in mice revealed that, TRPV4/p-eNOS expression and colocalization was reduced in endothelium in vivo. Finally, Ang II infusion induced vascular remodeling as evidenced by decreased lumen to wall ratio in resistant mesenteric arteries. These findings suggest that Ang II induces endothelial dysfunction and vascular remodeling via downregulation of TRPV4/eNOS pathway and may contribute to hypertension, independent of or in addition to its effect on vascular smooth muscle contraction.

6.
FASEB J ; 36(5): e22273, 2022 05.
Article in English | MEDLINE | ID: mdl-35349200

ABSTRACT

Mast cells (MCs) develop from hematopoietic progenitors and differentiate into mature MCs that reside within connective or mucosal tissues. Though the number of MCs in tissues usually remains constant, inflammation and asthma disturb this homeostasis, leading to proliferation of MCs. Understanding the signaling events behind this proliferative response could lead to the development of novel strategies for better management of allergic diseases. MC survival, proliferation, differentiation, and migration are all maintained by a MC growth factor, stem cell factor (SCF) via its receptor, KIT. Here, we explored how protein kinase C (PKC) redundancy influences MC proliferation in bone marrow-derived MC (BMMC). We found that SCF activates PKCα and PKCß isoforms, which in turn modulates KIT phosphorylation and internalization. Further, PKCα and PKCß activate p38 mitogen activated protein kinase (MAPK), and this axis subsequently regulates SCF-induced MC cell proliferation. To ascertain the individual roles of PKCα and PKCß, we knocked down either PKCα or PKCß or both via short hairpin RNA (shRNA) and analyzed KIT phosphorylation, p38 MAPK phosphorylation, and MC viability and proliferation. To our surprise, downregulation of neither PKCα nor PKCß affected MC viability and proliferation. In contrast, blocking both PKCα and PKCß significantly attenuated SCF-induced cell viability and proliferation, suggesting that PKCα and PKCß compensate for each other downstream of SCF signaling to enhance MC viability and proliferation. Our results not only suggest that PKC classical isoforms are novel therapeutic targets for SCF/MC-mediated inflammatory and allergic diseases, but they also emphasize the importance of inhibiting both PKCα and ß isoforms simultaneously to prevent MC proliferation.


Subject(s)
Mast Cells , Stem Cell Factor , Cell Proliferation , Cell Survival/physiology , Mast Cells/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Front Physiol ; 12: 756450, 2021.
Article in English | MEDLINE | ID: mdl-34867460

ABSTRACT

Endothelial permeability, leukocyte attachment, and unregulated oxidized LDL (oxLDL) uptake by macrophages leading to the formation of foam cells are all vital in the initiation and progression of atherosclerosis. During inflammation, several inflammatory mediators regulate this process through the expression of distinct oxLDL binding cell surface receptors on macrophages. We have previously shown that Leukotriene D4 (LTD4) promotes endothelial dysfunction, increasing endothelial permeability and enhancing TNFα-mediated attachment of monocytes to endothelium, which hints at its possible role in atherosclerosis. Here we analyzed the effect of LTD4 on macrophage function. Macrophages mainly express CysLT1R and flux calcium in response to LTD4. Further, LTD4 potentiates phagocytosis in macrophages as revealed by the uptake of zymosan particles. Notably, LTD4 augmented macrophage phagocytosis and oxLDL uptake which is sensitive to MK-571 [Montelukast (MK)], a CysLT1R-specific antagonist. Mechanistically, LTD4 upregulated two receptors central to foam cell formation, oxidized low-density lipoprotein receptor-1 (OLR1/LOX-1), and CD36 in a time and dose-dependent manner. Finally, LTD4 enhanced the secretion of chemokines MCP-1 and MIP1ß. Our results suggest that LTD4 contributes to atherosclerosis either through driving foam cell formation or recruitment of immune cells or both. CysLT1R antagonists are safely being used in the treatment of asthma, and the findings from the current study suggest that these can be re-purposed for the treatment of atherosclerosis.

8.
Cells ; 10(11)2021 11 06.
Article in English | MEDLINE | ID: mdl-34831281

ABSTRACT

Fibrosis is an irreversible, debilitating condition marked by the excessive production of extracellular matrix and tissue scarring that eventually results in organ failure and disease. Differentiation of fibroblasts to hypersecretory myofibroblasts is the key event in fibrosis. Although both soluble and mechanical factors are implicated in fibroblast differentiation, much of the focus is on TGF-ß signaling, but to date, there are no specific drugs available for the treatment of fibrosis. In this review, we describe the role for TRPV4 mechanotransduction in cardiac and lung fibrosis, and we propose TRPV4 as an alternative therapeutic target for fibrosis.


Subject(s)
Mechanotransduction, Cellular , TRPV Cation Channels/metabolism , Animals , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Humans , Myocardium/pathology , Signal Transduction
9.
Dyes Pigm ; 1942021 Oct.
Article in English | MEDLINE | ID: mdl-34366501

ABSTRACT

Near-infrared (NIR) emitting probes with very large Stokes' shifts play a crucial role in bioimaging applications, as the optical signals in this region exhibit high signal to background ratio and allow deeper tissue penetration. Herein we illustrate NIR-emitting probe 2 with very large Stokes' shifts (Δλ ≈ 260 - 272 nm) by integrating the excited-state intramolecular proton transfer (ESIPT) unit 2-(2'-hydroxyphenyl)benzoxazole (HBO) into a pyridinium derived cyanine. The ESIPT not only enhances the Stokes' shifts but also improves the quantum efficiency of the probe 2 (фfl = 0.27 - 0.40 in DCM). The application of 2 in live cells imaging reveals that compound 2 stains mitochondria in eukaryotic cells, normal human lungs fibroblast (NHLF), Zebrafish's neuromast hair cells, and support cells, and inner plasma membrane in prokaryotic cells, Escherichia coli (E. coli).

10.
Bone Rep ; 14: 100754, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33665237

ABSTRACT

Bone biomineralization is a complex process in which type I collagen and associated non-collagenous proteins (NCPs), including glycoproteins and proteoglycans, interact closely with inorganic calcium and phosphate ions to control the precipitation of nanosized, non-stoichiometric hydroxyapatite (HAP, idealized stoichiometry Ca10(PO4)6(OH)2) within the organic matrix of a tissue. The ability of certain vertebrate tissues to mineralize is critically related to several aspects of their function. The goal of this study was to identify specific NCPs in mineralizing and non-mineralizing tissues of two animal models, rat and turkey, and to determine whether some NCPs are unique to each type of tissue. The tissues investigated were rat femur (mineralizing) and tail tendon (non-mineralizing) and turkey leg tendon (having both mineralizing and non-mineralizing regions in the same individual specimen). An experimental approach ex vivo was designed for this investigation by combining sequential protein extraction with comprehensive protein mapping using proteomics and Western blotting. The extraction method enabled separation of various NCPs based on their association with either the extracellular organic collagenous matrix phases or the inorganic mineral phases of the tissues. The proteomics work generated a complete picture of NCPs in different tissues and animal species. Subsequently, Western blotting provided validation for some of the proteomics findings. The survey then yielded generalized results relevant to various protein families, rather than only individual NCPs. This study focused primarily on the NCPs belonging to the small leucine-rich proteoglycan (SLRP) family and the small integrin-binding ligand N-linked glycoproteins (SIBLINGs). SLRPs were found to be associated only with the collagenous matrix, a result suggesting that they are mainly involved in structural matrix organization and not in mineralization. SIBLINGs as well as matrix Gla (γ-carboxyglutamate) protein were strictly localized within the inorganic mineral phase of mineralizing tissues, a finding suggesting that their roles are limited to mineralization. The results from this study indicated that osteocalcin was closely involved in mineralization but did not preclude possible additional roles as a hormone. This report provides for the first time a spatial survey and comparison of NCPs from mineralizing and non-mineralizing tissues ex vivo and defines the proteome of turkey leg tendons as a model for vertebrate mineralization.

11.
Angiogenesis ; 24(3): 647-656, 2021 08.
Article in English | MEDLINE | ID: mdl-33656628

ABSTRACT

Transient receptor potential vanilloid 4 (TRPV4) is a ubiquitously expressed polymodally activated ion channel. TRPV4 has been implicated in tumor progression; however, the cell-specific role of TRPV4 in tumor growth, angiogenesis, and metastasis is unknown. Here, we generated endothelial-specific TRPV4 knockout (TRPV4ECKO) mice by crossing TRPV4lox/lox mice with Tie2-Cre mice. Tumor growth and metastasis were significantly increased in a syngeneic Lewis lung carcinoma tumor model of TRPV4ECKO mice compared to TRPV4lox/lox mice. Multiphoton microscopy, dextran leakage, and immunohistochemical analysis revealed increased tumor angiogenesis and metastasis that were correlated with aberrant leaky vessels (increased width and reduced pericyte and VE-cadherin coverage). Mechanistically, increases in VEGFR2, p-ERK, and MMP-9 expression and DQ gelatinase activity were observed in the TRPV4ECKO mouse tumors. Our results demonstrated that endothelial TRPV4 is a critical modulator of vascular integrity and tumor angiogenesis and that deletion of TRPV4 promotes tumor angiogenesis, growth, and metastasis.


Subject(s)
Carcinoma, Lewis Lung/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , TRPV Cation Channels/metabolism , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Mice , Mice, Knockout , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , TRPV Cation Channels/genetics
12.
Front Bioeng Biotechnol ; 9: 790489, 2021.
Article in English | MEDLINE | ID: mdl-35004649

ABSTRACT

Tumor angiogenesis is initiated and maintained by the tumor microenvironment through secretion of autocrine and paracrine factors, including extracellular vesicles (EVs). Although tumor-derived EVs (t-EVs) have been implicated in tumor angiogenesis, growth and metastasis, most studies on t-EVs are focused on proangiogenic miRNAs and growth factors. We have recently demonstrated that conditioned media from human lung tumor cells (A549) downregulate TRPV4 channels and transform normal endothelial cells to a tumor endothelial cell-like phenotype and induce abnormal angiogenesis in vitro, via t-EVs. However, the underlying molecular mechanism of t-EVs on endothelial cell phenotypic transition and abnormal angiogenesis in vivo remains unknown. Here, we demonstrate that t-EVs downregulate TRPV4 expression post-translationally and induce abnormal angiogenesis by activating Rho/Rho kinase/YAP/VEGFR2 pathways. Further, we demonstrate that t-EVs induce abnormal vessel formation in subcutaneously implanted Matrigel plugs in vivo (independent of tumors), which are characterized by increased VEGFR2 expression and reduced pericyte coverage. Taken together, our findings demonstrate that t-EVs induce abnormal angiogenesis via TRPV4 downregulation-mediated activation of Rho/Rho kinase/YAP/VEGFR2 pathways and suggest t-EVs and TRPV4 as novel targets for vascular normalization and cancer therapy.

13.
J Cell Physiol ; 236(5): 3770-3779, 2021 05.
Article in English | MEDLINE | ID: mdl-33078410

ABSTRACT

Transient receptor potential vanilloid 4 (TRPV4) channels are mechanosensitive ion channels that regulate systemic endothelial cell (EC) functions such as vasodilation, permeability, and angiogenesis. TRPV4 is expressed in retinal ganglion cells, Müller glia, pigment epithelium, microvascular ECs, and modulates cell volume regulation, calcium homeostasis, and survival. TRPV4-mediated physiological or pathological retinal angiogenesis remains poorly understood. Here, we demonstrate that TRPV4 is expressed, functional, and mechanosensitive in retinal ECs. The genetic deletion of TRPV4 did not affect postnatal developmental angiogenesis but increased pathological neovascularization in response to oxygen-induced retinopathy (OIR). Retinal vessels from TRPV4 knockout mice subjected to OIR exhibited neovascular tufts that projected into the vitreous humor and displayed reduced pericyte coverage compared with wild-type mice. These results suggest that TRPV4 is a regulator of retinal angiogenesis, its deletion augments pathological retinal angiogenesis, and that TRPV4 could be a novel target for the development of therapies against neovascular ocular diseases.


Subject(s)
Gene Deletion , Neovascularization, Physiologic , Retinal Neovascularization/metabolism , TRPV Cation Channels/metabolism , Animals , Endothelial Cells/metabolism , Humans , Mechanotransduction, Cellular , Mice, Inbred C57BL , Microvessels/pathology , Oxygen , Pericytes/pathology , Retina/pathology
14.
Exp Eye Res ; 201: 108257, 2020 12.
Article in English | MEDLINE | ID: mdl-32979394

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.


Subject(s)
Ciliary Body/metabolism , Lens, Crystalline/metabolism , Retina/metabolism , TRPV Cation Channels/metabolism , Animals , Cell Membrane/metabolism , Humans , Osmotic Pressure , Signal Transduction
15.
Sci Rep ; 10(1): 9827, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32555397

ABSTRACT

Asthma is characterized by pathological airway remodeling resulting from persistent myofibroblast activation. Although transforming growth factor beta 1 (TGFß1), mechanical signals, and reactive oxygen species (ROS) are implicated in fibroblast differentiation, their integration is still elusive. We identified that Transient Receptor Potential Vanilloid 4 (TRPV4), a mechanosensitive ion channel mediates lung fibroblast (LF) differentiation and D. farinae-induced airway remodeling via a novel TRPV4-NADPH Oxidase 4 (NOX4) interaction. NOX4-mediated ROS production is essential for TGFß1-induced LF differentiation via myocardin-related transcription factor-A (MRTF-A) and plasminogen activator inhibitor 1 (PAI-1). Importantly, TRPV4 inhibition prevented TGFß1-induced NOX4 expression and ROS production. Both TRPV4 and NOX4 are activated by phosphatidylinositol 3-kinase (PI3K) downstream of TGFß1, and signals from both TRPV4 and Rac are necessary for NOX4 upregulation. Notably, NOX4 expression is higher in fibroblasts derived from asthmatic patients (disease human LF; DHLF) in comparison to non-asthmatics (normal human LF; NHLF). Further, NOX4 expression is up-regulated in the lungs of D.farinae-treated wild type mice (WT) relative to saline-treated WT, which was attenuated in TRPV4 knockout (KO) mice. Our findings suggest that TRPV4 integrates TGFß1 and ROS signaling through NOX4 and, TRPV4-NOX4 interaction is amenable to target lung remodeling during asthma.


Subject(s)
Airway Remodeling , Cell Differentiation , Fibroblasts/cytology , NADPH Oxidase 4/metabolism , TRPV Cation Channels/metabolism , Animals , Gene Expression Regulation , Gene Knockout Techniques , Humans , Mice , NADPH Oxidase 4/deficiency , NADPH Oxidase 4/genetics , Oxidation-Reduction , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
16.
Basic Res Cardiol ; 115(2): 14, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31925567

ABSTRACT

Cardiac fibrosis caused by adverse cardiac remodeling following myocardial infarction can eventually lead to heart failure. Although the role of soluble factors such as TGF-ß is well studied in cardiac fibrosis following myocardial injury, the physiological role of mechanotransduction is not fully understood. Here, we investigated the molecular mechanism and functional role of TRPV4 mechanotransduction in cardiac fibrosis. TRPV4KO mice, 8 weeks following myocardial infarction (MI), exhibited preserved cardiac function compared to WT mice. Histological analysis demonstrated reduced cardiac fibrosis in TRPV4KO mice. We found that WT CF exhibited hypotonicity-induced calcium influx and extracellular matrix (ECM)-stiffness-dependent differentiation in response to TGF-ß1. In contrast, TRPV4KO CF did not display hypotonicity-induced calcium influx and failed to differentiate on high-stiffness ECM gels even in the presence of saturating amounts of TGF-ß1. Mechanistically, TRPV4 mediated cardiac fibrotic gene promoter activity and fibroblast differentiation through the activation of the Rho/Rho kinase pathway and the mechanosensitive transcription factor MRTF-A. Our findings suggest that genetic deletion of TRPV4 channels protects heart from adverse cardiac remodeling following MI by modulating Rho/MRTF-A pathway-mediated cardiac fibroblast differentiation and cardiac fibrosis.


Subject(s)
Cell Differentiation , Fibroblasts/metabolism , Gene Deletion , Myocardial Infarction/prevention & control , Myocardium/metabolism , TRPV Cation Channels/deficiency , Ventricular Remodeling , Animals , Calcium Signaling , Cells, Cultured , Disease Models, Animal , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibroblasts/pathology , Fibrosis , Mechanotransduction, Cellular , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , TRPV Cation Channels/genetics , Trans-Activators/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
17.
Chem Commun (Camb) ; 55(88): 13223-13226, 2019 Oct 31.
Article in English | MEDLINE | ID: mdl-31595909

ABSTRACT

There are a limited number of near-infrared (NIR) emitting (λem = 700-900 nm) molecular probes for imaging applications. A NIR-emitting probe that exhibits emission at ∼800 nm with a large Stokes shift was synthesized and found to exhibit excellent selectivity towards mitochondria for live-cell imaging. The photophysical properties were attributed to an excited "cyanine structure" via intramolecular charge transfer (ICT) involving a phenol group.


Subject(s)
Carbocyanines/chemistry , Fibroblasts/chemistry , Fluorescent Dyes/chemistry , Oligodendroglia/chemistry , Optical Imaging , Phenols/chemistry , Cell Line , Humans , Infrared Rays , Lung/cytology , Molecular Structure , Spectrometry, Fluorescence
18.
ACS Appl Bio Mater ; 2(9): 4037-4043, 2019 Sep 16.
Article in English | MEDLINE | ID: mdl-35021337

ABSTRACT

Lysosome imaging without perturbing intracellular activity remains challenging, as the current commercial lysosome probes contain weakly basic amino groups that could perturb lysosome pH. Herein, we illustrate NIR-emitting dyes 2 and 3 (λem ≈ 700 nm) with very large Stokes' shifts (Δλ = 231-246 nm), attributing to the presence of a 2-hydroxyphenyl(benzo[d]oxazol) (HBO) unit that undergoes excited-state intramolecular proton transfer (ESIPT). The structures of 2 and 3 also contain a hemicyanine unit with benzothiazolium and indolium as a terminal group, respectively. Although the fluorescent probe 2 (Φfl ≈ 0.28-0.35 in CH2Cl2) does not contain any basic amino functional group, it exhibits excellent selectivity for staining intracellular lysosomes, showing the potential for long-term in vivo lysosome imaging without "alkalinizing effect." However, probe 3 (Φfl ≈ 0.27, in CH2Cl2) exhibits excellent selectivity toward mitochondria. The observation showed that the terminal group in the hemicyanine unit played an essential role in guiding the intracellular selectivity to different organelles. In addition, the probes also displayed a transparent optical window between 520 and 590 nm, which is useful to achieve multicolor co-staining study, without fluorescence crosstalk that is a common problem on fluorescence microscopes.

19.
Front Cell Dev Biol ; 7: 344, 2019.
Article in English | MEDLINE | ID: mdl-31921855

ABSTRACT

The soluble and mechanical microenvironment surrounding endothelial cells influences and instructs them to form new blood vessels. The cells in the pathological tumor microenvironment release extracellular vesicles (EVs) for paracrine signaling. EVs have been shown to induce angiogenesis by communicating with endothelial cells, but the underlying molecular mechanisms are not well known. We have recently shown that the mechanosensitive ion channel transient receptor vanilloid 4 (TRPV4) expression and activity is significantly reduced in tumor endothelial cells (TEC), and that activation of TRPV4 normalized the tumor vasculature and improved cancer therapy. However, whether and how the tumor microenvironment downregulates TRPV4 and transforms the normal endothelial cell phenotype remains unknown. To explore this, we exposed normal human endothelial cells (hNEC) to human lung tumor cell conditioned media (TCM) and measured phenotypic changes and angiogenesis. We found that treatment with TCM transformed hNEC to a TEC-like phenotype (hTEC) as evidenced by increased expression of tumor endothelial cell marker 8 (TEM8) and exhibition of abnormal angiogenesis on 2D-Matrigels compared to normal hNEC. Mechanistically, expression and activity of TRPV4 was decreased in hTEC. Further, when pre-treated with exosome inhibitor GW4869, TCM failed to induce hNEC transformation to hTEC. Finally, addition of purified EVs from TCM induced transformation of hNEC to hTEC as evidenced by abnormal angiogenesis in vitro. Taken together, our results suggest that the pathological (tumor) microenvironment transforms normal endothelial cells into a tumor endothelial cell-like phenotype through EVs via the downregulation of TRPV4.

20.
Cancer Lett ; 442: 15-20, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30401632

ABSTRACT

The transient receptor potential vanilloid 4 (TRPV4) channel is a mechanosensor in endothelial cells (EC) that regulates cyclic strain-induced reorientation and flow-mediated nitric oxide production. We have recently demonstrated that TRPV4 expression is reduced in tumor EC and tumors grown in TRPV4KO mice exhibited enhanced growth and immature leaky vessels. However, the mechanism by which TRPV4 regulates tumor vascular integrity and metastasis is not known. Here, we demonstrate that VE-cadherin expression at the cell-cell contacts is significantly reduced in TRPV4-deficient tumor EC and TRPV4KO EC. In vivo angiogenesis assays with Matrigel of varying stiffness (700-900 Pa) revealed a significant stiffness-dependent reduction in VE-cadherin-positive vessels in Matrigel plugs from TRPV4KO mice compared with WT mice, despite an increase in vessel growth. Further, syngeneic Lewis Lung Carcinomatumor experiments demonstrated a significant decrease in VE-cadherin positive vessels in TRPV4KO tumors compared with WT. Functionally, enhanced tumor cell metastasis to the lung was observed in TRPV4KO mice. Our findings demonstrate that TRPV4 channels regulate tumor vessel integrity by maintaining VE-cadherin expression at cell-cell contacts and identifies TRPV4 as a novel target for metastasis.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Carcinoma, Lewis Lung/blood supply , Cell Movement , Endothelial Cells/metabolism , Intercellular Junctions/metabolism , Lung Neoplasms/blood supply , Mechanotransduction, Cellular , Neovascularization, Pathologic , TRPV Cation Channels/metabolism , Animals , Antigens, CD/genetics , Cadherins/genetics , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/secondary , Endothelial Cells/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Intercellular Junctions/genetics , Intercellular Junctions/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...