Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Compr Physiol ; 14(2): 5389-5406, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-39109978

ABSTRACT

Uncontrolled angiogenesis underlies various pathological conditions such as cancer, age-related macular degeneration (AMD), and proliferative diabetic retinopathy (PDR). Hence, targeting pathological angiogenesis has become a promising strategy for the treatment of cancer and neovascular ocular diseases. However, current pharmacological treatments that target VEGF signaling have met with limited success either due to acquiring resistance against anti-VEGF therapies with serious side effects including nephrotoxicity and cardiovascular-related adverse effects in cancer patients or retinal vasculitis and intraocular inflammation after intravitreal injection in patients with AMD or PDR. Therefore, there is an urgent need to develop novel strategies which can control multiple aspects of the pathological microenvironment and regulate the process of abnormal angiogenesis. To this end, vascular normalization has been proposed as an alternative for antiangiogenesis approach; however, these strategies still focus on targeting VEGF or FGF or PDGF which has shown adverse effects. In addition to these growth factors, calcium has been recently implicated as an important modulator of tumor angiogenesis. This article provides an overview on the role of major calcium channels in endothelium, TRP channels, with a special focus on TRPV4 and its downstream signaling pathways in the regulation of pathological angiogenesis and vascular normalization. We also highlight recent findings on the modulation of TRPV4 activity and endothelial phenotypic transformation by tumor microenvironment through Rho/YAP/VEGFR2 mechanotranscriptional pathways. Finally, we provide perspective on endothelial TRPV4 as a novel VEGF alternative therapeutic target for vascular normalization and improved therapy. © 2024 American Physiological Society. Compr Physiol 14:5389-5406, 2024.


Subject(s)
Neovascularization, Pathologic , Humans , Animals , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/physiology , Signal Transduction
2.
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
3.
BMJ Open ; 12(4): e049119, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35487526

ABSTRACT

INTRODUCTION: The standard of care for patients with localised rectal cancer is radical surgery, often combined with preoperative neoadjuvant (chemo)radiotherapy. While oncologically effective, this treatment strategy is associated with operative mortality risks, significant morbidity and stoma formation. An alternative approach is chemoradiotherapy to try to achieve a sustained clinical complete response (cCR). This non-surgical management can be attractive, particularly for patients at high risk of surgical complications. Modern radiotherapy techniques allow increased treatment conformality, enabling increased radiation dose to the tumour while reducing dose to normal tissue. The objective of this trial is to assess if radiotherapy dose escalation increases the cCR rate, with acceptable toxicity, for treatment of patients with early rectal cancer unsuitable for radical surgery. METHODS AND ANALYSIS: APHRODITE (A Phase II trial of Higher RadiOtherapy Dose In The Eradication of early rectal cancer) is a multicentre, open-label randomised controlled phase II trial aiming to recruit 104 participants from 10 to 12 UK sites. Participants will be allocated with a 2:1 ratio of intervention:control. The intervention is escalated dose radiotherapy (62 Gy to primary tumour, 50.4 Gy to surrounding mesorectum in 28 fractions) using simultaneous integrated boost. The control arm will receive 50.4 Gy to the primary tumour and surrounding mesorectum. Both arms will use intensity-modulated radiotherapy and daily image guidance, combined with concurrent chemotherapy (capecitabine, 5-fluorouracil/leucovorin or omitted). The primary endpoint is the proportion of participants with cCR at 6 months after start of treatment. Secondary outcomes include early and late toxicities, time to stoma formation, overall survival and patient-reported outcomes (European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaires QLQ-C30 and QLQ-CR29, low anterior resection syndrome (LARS) questionnaire). ETHICS AND DISSEMINATION: The trial obtained ethical approval from North West Greater Manchester East Research Ethics Committee (reference number 19/NW/0565) and is funded by Yorkshire Cancer Research. The final trial results will be published in peer-reviewed journals and adhere to International Committee of Medical Journal Editors guidelines. TRIAL REGISTRATION NUMBER: ISRCTN16158514.


Subject(s)
Rectal Neoplasms , Chemoradiotherapy/adverse effects , Chemoradiotherapy/methods , Clinical Trials, Phase II as Topic , Humans , Multicenter Studies as Topic , Postoperative Complications , Quality of Life , Randomized Controlled Trials as Topic , Rectal Neoplasms/radiotherapy , Syndrome
4.
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
5.
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
6.
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.

7.
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
8.
Ulster Med J ; 89(1): 25-28, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32218624

ABSTRACT

INTRODUCTION: Acute appendicitis is a common surgical emergency, with a prevalence of 112 per 100,000 people per year in Europe. Negative appendicectomy is defined as a pathologically normal appendix removed from patient suspected with appendicitis. Negative appendectomy rate (NAR) has been reported to be around 15-25%. We aimed to evaluate our unit's negative appendectomy rate and the effect of pre-operative imaging on NAR. METHOD: A retrospective study including all patients who underwent both open and laparoscopic emergency appendicectomy in a single district general hospital from 2017-2018. Clinical information including cost was calculated based on the 2017/18 national tariff payment system. Patients under 18 years old were excluded from this study. RESULTS: Two hundred thirty-two patients were included in this study, of which 69 (29.74%) had a pre-operative CT scan. The mean length of stay was 2.57 days. The sensitivity, specificity, positive predictive value and negative predictive value for CT were 77.8%, 100%, 87.5% and 100%. The negative appendicectomy rate with and without pre-operative CT scan were 7.25% and 22.09% respectively. Based on the 2017/18 national tariff payment system, a CT abdomen and pelvis with contrast and emergency appendicectomy with CC score of 0 cost 92 and 2370 pounds respectively. The total cost of patients who underwent appendicectomy without imaging was £ 322,320. If all patients undergo pre-operative CT, with a reduction of 15% in negative appendicectomy rate, the overall total cost would significantly lower to £ 36,212. CONCLUSION: Our study demonstrated that the negative appendicectomy rate could be improved by preoperative imaging. The study also showed that implementation of preoperative imaging for suspected appendicitis cases could save costs, allowing better allocation of resources.


Subject(s)
Appendectomy/statistics & numerical data , Appendicitis/diagnostic imaging , Unnecessary Procedures , Appendectomy/economics , Humans , Predictive Value of Tests , Preoperative Care , Retrospective Studies , Sensitivity and Specificity , Tomography, X-Ray Computed , United Kingdom
9.
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
10.
Proc Natl Acad Sci U S A ; 116(1): 199-204, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30559191

ABSTRACT

Cysteinyl leukotrienes (cys-LTs) are proinflammatory mediators that enhance vascular permeability through distinct receptors (CysLTRs). We found that CysLT2R regulates angiogenesis in isolated mouse endothelial cells (ECs) and in Matrigel implants in WT mice and enhances EC contraction and permeability via the Rho-dependent myosin light chain 2 and vascular endothelial (VE)-cadherin axis. Since solid tumors utilize aberrant angiogenesis for their growth and metastasis and their vessels exhibit vascular hyperpermeability, we hypothesized that CysLT2R, via its actions on the endothelium, might regulate tumor growth. Both tumor growth and metastases of adoptively transferred syngeneic Lewis lung carcinoma (LLC) cells are significantly reduced in CysLT2R-null mice (Cysltr2-/-) compared with WT and CysLT1R-null mice (Cysltr1-/-). In WT recipients of LLC cells, CysLT2R expression is significantly increased in the tumor vasculature, compared with CysLT1R. Further, the tumor vasculature in Cysltr2-/- recipients exhibited significantly improved integrity, as revealed by increased pericyte coverage and decreased leakage of i.v.-administered Texas Red-conjugated dextran. Administration of a selective CysLT2R antagonist significantly reduced LLC tumor volume, vessel density, dextran leakage, and metastases in WT mice, highlighting CysLT2R as a VEGF-independent regulator of the vasculature promoting risk of metastasis. Thus, both genetic and pharmacological findings establish CysLT2R as a gateway for angiogenesis and EC dysregulation in vitro and ex vivo and in an in vivo model with a mouse tumor. Our data suggest CysLT2R as a possible target for intervention.


Subject(s)
Endothelial Cells/drug effects , Neovascularization, Pathologic/chemically induced , Receptors, Leukotriene/metabolism , Animals , Capillary Permeability/drug effects , Cyclohexanecarboxylic Acids/pharmacology , Gene Knockout Techniques , Leukotriene Antagonists/pharmacology , Lung Neoplasms/drug therapy , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/drug therapy , Neoplasm Transplantation , Neoplasms, Experimental , Neovascularization, Pathologic/drug therapy , Phthalic Acids/pharmacology , Receptors, Leukotriene/drug effects
11.
FASEB J ; 33(1): 195-203, 2019 01.
Article in English | MEDLINE | ID: mdl-29957061

ABSTRACT

VEGF signaling via VEGF receptor-2 (VEGFR2) is a major regulator of endothelial cell (EC) functions, including angiogenesis. Although most studies of angiogenesis focus on soluble VEGF signaling, mechanical signaling also plays a critical role. Here, we examined the consequence of disruption of mechanical signaling on soluble signaling pathways. Specifically, we observed that small interfering RNA (siRNA) knockdown of a mechanosensitive ion channel, transient receptor potential vanilloid 4 (TRPV4), significantly reduced perinuclear (Golgi) VEGFR2 in human ECs with a concomitant increase in phosphorylation at Y1175 and membrane translocation. TRPV4 knockout (KO) ECs exhibited increased plasma membrane localization of phospho-VEGFR2 compared with normal ECs. The knockdown also increased phospho-VEGFR2 in whole cell lysates and membrane fractions compared with control siRNA-treated cells. siRNA knockdown of TRPV4 enhanced nuclear localization of mechanosensitive transcription factors, yes-associated protein/transcriptional coactivator with PDZ-binding motif via rho kinase, which were shown to increase VEGFR2 trafficking to the plasma membrane. Furthermore, TRPV4 deletion/knockdown enhanced VEGF-mediated migration in vitro and increased expression of VEGFR2 in vivo in the vasculature of TRPV4 KO tumors compared with wild-type tumors. Our results thus show that TRPV4 channels regulate VEGFR2 trafficking and activation to identify novel cross-talk between mechanical (TRPV4) and soluble (VEGF) signaling that controls EC migration and angiogenesis.-Kanugula, A. K., Adapala, R. K., Midha, P., Cappelli, H. C., Meszaros, J. G., Paruchuri, S., Chilian, W. M., Thodeti, C. K., Novel noncanonical regulation of soluble VEGF/VEGFR2 signaling by mechanosensitive ion channel TRPV4.


Subject(s)
Carcinoma, Lewis Lung/pathology , Cell Movement , Endothelium, Vascular/pathology , Mechanotransduction, Cellular , TRPV Cation Channels/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Cell Proliferation , Endothelium, Vascular/metabolism , Humans , Mice , Phosphorylation , Signal Transduction , TRPV Cation Channels/genetics , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
12.
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
13.
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.

14.
Oncotarget ; 7(18): 25849-61, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27029071

ABSTRACT

Targeting angiogenesis is considered a promising therapy for cancer. Besides curtailing soluble factor mediated tumor angiogenesis, understanding the unexplored regulation of angiogenesis by mechanical cues may lead to the identification of novel therapeutic targets. We have recently shown that expression and activity of mechanosensitive ion channel transient receptor potential vanilloid 4 (TRPV4) is suppressed in tumor endothelial cells and restoring TRPV4 expression or activation induces vascular normalization and improves cancer therapy. However, the molecular mechanism(s) by which TRPV4 modulates angiogenesis are still in their infancy. To explore how TRPV4 regulates angiogenesis, we have employed TRPV4 null endothelial cells (TRPV4KO EC) and TRPV4KO mice. We found that absence of TRPV4 (TRPV4KO EC) resulted in a significant increase in proliferation, migration, and abnormal tube formation in vitro when compared to WT EC. Concomitantly, sprouting angiogenesis ex vivo and vascular growth in vivo was enhanced in TRPV4KO mice. Mechanistically, we observed that loss of TRPV4 leads to a significant increase in basal Rho activity in TRPV4KO EC that corresponded to their aberrant mechanosensitivity on varying stiffness ECM gels. Importantly, pharmacological inhibition of the Rho/Rho kinase pathway by Y-27632 normalized abnormal mechanosensitivity and angiogenesis exhibited by TRPV4KO EC in vitro. Finally, Y-27632 treatment increased pericyte coverage and in conjunction with Cisplatin, significantly reduced tumor growth in TRPV4KO mice. Taken together, these data suggest that TRPV4 regulates angiogenesis endogenously via modulation of EC mechanosensitivity through the Rho/Rho kinase pathway and can serve as a potential therapeutic target for cancer therapy.


Subject(s)
Carcinoma, Lewis Lung/pathology , Neovascularization, Pathologic/metabolism , TRPV Cation Channels/metabolism , rho-Associated Kinases/metabolism , Animals , Carcinoma, Lewis Lung/metabolism , Cell Movement/physiology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/pathology
15.
Sci Rep ; 5: 14257, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26388427

ABSTRACT

Endothelial cell proliferation is a critical event during angiogenesis, regulated by both soluble factors and mechanical forces. Although the proliferation of tumor cells is studied extensively, little is known about the proliferation of tumor endothelial cells (TEC) and its contribution to tumor angiogenesis. We have recently shown that reduced expression of the mechanosensitive ion channel TRPV4 in TEC causes aberrant mechanosensitivity that result in abnormal angiogenesis. Here, we show that TEC display increased proliferation compared to normal endothelial cells (NEC). Further, we found that TEC exhibit high basal ERK1/2 phosphorylation and increased expression of proliferative genes important in the G1/S phase of the cell cycle. Importantly, pharmacological activation of TRPV4, with a small molecular activator GSK1016790A (GSK), significantly inhibited TEC proliferation, but had no effect on the proliferation of NEC or the tumor cells (epithelial) themselves. This reduction in TEC proliferation by TRPV4 activation was correlated with a decrease in high basal ERK1/2 phosphorylation. Finally, using a syngeneic tumor model revealed that TRPV4 activation, with GSK, significantly reduced endothelial cell proliferation in vivo. Our findings suggest that TRPV4 channels regulate tumor angiogenesis by selectively inhibiting tumor endothelial cell proliferation.


Subject(s)
Neoplasms/metabolism , TRPV Cation Channels/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Leucine/analogs & derivatives , Leucine/pharmacology , MAP Kinase Signaling System , Mice , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Sulfonamides/pharmacology , TRPV Cation Channels/drug effects , TRPV Cation Channels/genetics , Up-Regulation
16.
Can J Physiol Pharmacol ; 92(7): 598-604, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24959995

ABSTRACT

Diabetes is an independent risk factor for cardiovascular disease that can eventually cause cardiomyopathy and heart failure. Cardiac fibroblasts (CF) are the critical mediators of physiological and pathological cardiac remodeling; however, the effects of hyperglycemia on cardiac fibroblast function and differentiation is not well known. Here, we performed a comprehensive investigation on the effects of hyperglycemia on cardiac fibroblasts and show that hyperglycemia enhances cardiac fibroblast function and differentiation. We found that high glucose treatment increased collagen I, III, and VI gene expression in rat adult cardiac fibroblasts. Interestingly, hyperglycemia increased CF migration and proliferation that is augmented by collagen I and III. Surprisingly, we found that short term hyperglycemia transiently inhibited ERK1/2 activation but increased AKT phosphorylation. Finally, high glucose treatment increased spontaneous differentiation of cardiac fibroblasts to myofibroblasts with increasing passage compared with low glucose. Taken together, these findings suggest that hyperglycemia induces cardiac fibrosis by modulating collagen expression, migration, proliferation, and differentiation of cardiac fibroblasts.


Subject(s)
Cell Differentiation , Fibroblasts/metabolism , Hyperglycemia/metabolism , Myocardium/metabolism , Animals , Blood Glucose/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Collagen/metabolism , Fibroblasts/pathology , Fibrosis , Hyperglycemia/pathology , Male , Myocardium/pathology , Rats, Sprague-Dawley , Signal Transduction
17.
Sci Rep ; 3: 3274, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24253666

ABSTRACT

Cysteinyl leukotrienes (cys-LTs), LTC4, LTD4, LTE4 are potent inflammatory lipid mediators that act through two distinct G-protein-coupled receptors, CysLT1R and CysLT2R. Although cys-LTs are shown to induce vascular leakage and atherosclerosis, the molecular mechanism by which cys-LTs modulate endothelial function is not known. Here, we show that cys-LTs (LTC4 and LTD4) induce robust calcium influx in human umbilical vein endothelial cells (HUVECs) through CysLT2R, but not CysLT1R. Further, cys-LT treatment induced endothelial cell (EC) contraction leading to monolayer disruption via CysLT2R/Rho kinase dependent pathway. Furthermore, stimulation with cys-LTs potentiated TNFα-induced VCAM-1 expression and leukocyte recruitment to ECs through CysLT2R. In contrast, we found that both LTC4 and LTD4 stimulated EC proliferation through CysLT1R. Taken together, these results suggest that cys-LTs induce endothelial inflammation and proliferation via CysLT2R/Rho kinase and CysLT1R/Erk dependent pathways, respectively, which play critical role in the etiology of cardiovascular diseases such as atherosclerosis and myocardial infarction.


Subject(s)
Cysteine/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Leukotrienes/pharmacology , Receptors, Leukotriene/metabolism , Signal Transduction/drug effects , Calcium/metabolism , Calcium Signaling , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Tumor Necrosis Factor-alpha/pharmacology , rho-Associated Kinases/metabolism
18.
J Biomed Mater Res A ; 101(5): 1502-10, 2013 May.
Article in English | MEDLINE | ID: mdl-23184520

ABSTRACT

Previously, we used 2D films to identify an annealed PCL-PDLLA phase-separated blend morphology which provided nanoscale surface texture and patterning that stimulated osteoblast differentiation. In order to translate these 2D surface nanopatterning effects to the walls of 3D salt-leached scaffolds, the blend phase morphology of scaffold walls must be characterized. For salt-leached scaffolds, NaCl is used as a porogen, which may affect phase separation in PCL-PDLLA blends. However, it is not possible to characterize the surface blend morphology of 3D scaffold walls using standard approaches such as AFM or optical microscopy, since scaffolds are too rough for AFM and do not transmit light for optical microscopy. We introduce a 2.5D approach that mimics the processing conditions of 3D salt-leached scaffolds, but has a geometry amenable to surface characterization by AFM and optical microscopy. For the 2.5D approach, PCL-PDLLA blend films were covered with NaCl crystals prior to annealing. The presence of NaCl significantly influenced blend morphology in PCL-PDLLA 2.5D constructs causing increased surface roughness, higher percent PCL area on the surface and a smaller PCL domain size. During cell culture on 2.5D constructs, osteoblast (MC3T3-E1) and dermal endothelial cell (MDEC) adhesion were enhanced on PCL-PDLLA blends that were annealed with NaCl while chondrogenic cell (ATDC5) adhesion was diminished. This work introduces a 2.5D approach that mimicked 3D salt-leached scaffold processing, but enabled characterization of scaffold surface properties by AFM and light microscopy, to demonstrate that the presence of NaCl during annealing strongly influenced polymer blend surface morphology and cell adhesion.


Subject(s)
Polyesters/chemistry , Sodium Chloride/chemistry , Tissue Scaffolds/chemistry , 3T3 Cells , Animals , Cell Adhesion , Cell Count , Cell Line , Chondrocytes/cytology , Endothelial Cells/cytology , Hot Temperature , Mice , Osteoblasts/cytology , Surface Properties , Tissue Engineering
19.
J Mol Cell Cardiol ; 54: 45-52, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23142541

ABSTRACT

The phenotypic switch underlying the differentiation of cardiac fibroblasts into hypersecretory myofibroblasts is critical for cardiac remodeling following myocardial infarction. Myofibroblasts facilitate wound repair in the myocardium by secreting and organizing extracellular matrix (ECM) during the wound healing process. However, the molecular mechanisms involved in myofibroblast differentiation are not well known. TGF-ß has been shown to promote differentiation and this, combined with the robust mechanical environment in the heart, lead us to hypothesize that the mechanotransduction and TGF-ß signaling pathways play active roles in the differentiation of cardiac fibroblasts to myofibroblasts. Here, we show that the mechanosensitve ion channel TRPV4 is required for TGF-ß1-induced differentiation of cardiac fibroblasts into myofibroblasts. We found that the TRPV4-specific antagonist AB159908 and siRNA knockdown of TRPV4 significantly inhibited TGFß1-induced differentiation as measured by incorporation of α-SMA into stress fibers. Further, we found that TGF-ß1-induced myofibroblast differentiation was dependent on ECM stiffness, a response that was attenuated by TRPV4 blockade. Finally, TGF-ß1 treated fibroblasts exhibited enhanced TRPV4 expression and TRPV4-mediated calcium influx compared to untreated controls. Taken together these results suggest for the first time that the mechanosensitive ion channel, TRPV4, regulates cardiac fibroblast differentiation to myofibroblasts by integrating signals from TGF-ß1 and mechanical factors.


Subject(s)
Calcium Signaling , Cell Differentiation , Fibroblasts/physiology , Mechanotransduction, Cellular , TRPV Cation Channels/metabolism , Animals , Cymenes , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Gene Knockdown Techniques , Male , Monoterpenes/pharmacology , Myocardium/cytology , Myofibroblasts/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/metabolism , TRPV Cation Channels/genetics , Transforming Growth Factor beta1/physiology
20.
Am J Physiol Cell Physiol ; 303(2): C179-91, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22592401

ABSTRACT

Hyperleptinemia, characteristic of diabetes and a hallmark feature of human obesity, contributes to the increased risk of atherosclerotic complications. However, molecular mechanisms mediating leptin-induced atherogenesis and gene expression in vascular cells remain incompletely understood. Accumulating evidence documents a critical role of a potent antiangiogenic and proatherogenic matricellular protein, thrombospondin-1 (TSP-1), in atherosclerosis. Although previous studies reported elevated TSP-1 levels in both diabetic and obese patients and rodent models, there is no direct information on TSP-1 expression in vascular cells in response to leptin. In the present study, we show that leptin upregulates TSP-1 expression in cultured human aortic smooth muscle cells (HASMC) in vitro, and this increase occurs at the level of transcription, revealed by mRNA stability and TSP-1 promoter-reporter assays. Utilizing specific pharmacological inhibitors and siRNA approaches, we demonstrate that upregulation of TSP-1 expression by leptin is mediated by JAK2/ERK/JNK-dependent mechanisms. Furthermore, we report that while ERK and JNK are required for both the constitutive and leptin-induced expression of TSP-1, JAK-2 appears to be specifically involved in leptin-mediated TSP-1 upregulation. Finally, we found that increased HASMC migration and proliferation in response to leptin is significantly inhibited by a TSP-1 blocking antibody, thereby revealing the physiological significance of leptin-TSP-1 crosstalk. Taken together, these findings demonstrate, for the first time, that leptin has a direct regulatory effect on TSP-1 expression in HASMCs, underscoring a novel role of TSP-1 in hyperleptinemia-induced atherosclerotic complications.


Subject(s)
Janus Kinase 2/biosynthesis , Leptin/physiology , MAP Kinase Signaling System/physiology , Muscle, Smooth, Vascular/metabolism , Thrombospondin 1/biosynthesis , Up-Regulation/physiology , Animals , Cell Movement/physiology , Cells, Cultured , Gene Expression Regulation/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL