Figure 1 - uploaded by Linley E Watson
Content may be subject to copyright.
beta 1 -Integrin Receptors and Ligands. Based on their association in heterodimers, the integrin family can be divided in the beta 1 , alpha V and beta 2 subgroups. beta 1 and alpha V members are ubiquitously expressed; the beta 2 subgroup is selectively expressed in leukocytes. Bars connecting alpha and beta subunits indicate known heterodimers. Ligands for each integrin 

beta 1 -Integrin Receptors and Ligands. Based on their association in heterodimers, the integrin family can be divided in the beta 1 , alpha V and beta 2 subgroups. beta 1 and alpha V members are ubiquitously expressed; the beta 2 subgroup is selectively expressed in leukocytes. Bars connecting alpha and beta subunits indicate known heterodimers. Ligands for each integrin 

Source publication
Article
Full-text available
Integrins are heterodimeric cell-surface molecules, which act as the principle mediators of molecular dialog between a cell and its extracellular matrix environment. In addition to their structural functions, integrins mediate signaling from the extracellular space into the cell through integrin-associated signaling and adaptor molecules such as FA...

Context in source publication

Context 1
... are heterodimeric cell-surface molecules, which act as the principle mediators of molecular dialog between a cell and its extracellular matrix environment. In addition to their structural functions, integrins mediate signaling from the extracellular space into the cell through integrin-associated signaling and adaptor molecules such as FAK (focal adhesion kinase), ILK (integrin-linked kinase), PINCH (particularly interesting new cysteine-histidine rich protein) and Nck2 (non-catalytic (region of) tyrosine kinase adaptor protein- 2). Via these molecules, integrin signaling tightly and cooperatively interacts with receptor tyrosine kinases (RTKs) signaling to regulate survival, proliferation and cell shape as well as polarity, adhesion, migration and differentiation. In the heart and blood vessels, the function and regulation of these molecules can be partially disturbed and thus contribute to cardiovascular diseases such as cardiac hypertrophy and atherosclerosis. In this review, we discuss the primary mechanisms of action and signaling of integrins in the cardiac and vascular system in normal and pathological states, as well as therapeutic strategies for targeting these systems (1). Integrins are a superfamily of heterodimeric cell surface receptors involved in cell–cell and cell–matrix adhesion (2). In addition to providing a direct link between the extracellular matrix (ECM) and cytoskeleton, integrins also regulate the production of second messengers within the cell. As multi-functional molecules, integrins are involved in organogenesis, anchoring of stem cells to niches, regulation of gene expression, cell proliferation, differentiation, migration and death (2-8). An important function of integrins is the ability to convert mechanical forces into biochemical signals (7, 9-18). Because dysregulation of integrins is involved in the pathogenesis of several disease states including atherosclerosis and cardiac hypertrophy (5, 19), extensive efforts have been directed toward understanding how integrins couple to signal transduction systems and integrate with other receptor systems. Cell adhesion molecules of the integrin family consist of 18 alpha and 8 beta subunits, which form 24 known alpha-beta-heterodimers depending on cell type and cellular function. Each integrin subunit has a large extracellular, a short transmembrane and small intracellular domain with a total of >1600 amino acids. Integrins are the main receptors for extracellular matrix proteins like collagen, fibronectin and laminin (Figure 1). The alpha subunit generally confers ligand specificity (20, 21) and the beta subunit is important for interacting with the cytoplasmic environment. Cell–matrix interaction via integrins is essential for embryonic development, as well as proliferation, survival, adhesion, differentiation and migration of cells. Ligand binding to the extracellular integrin domain induces conformational changes and integrin clustering for activation of signaling cascades and recruitment of multiprotein complexes to focal adhesions (3, 9). Integrins transmit signals through a variety of intracellular protein kinases and adaptor molecules such as ILK, FAK, talin, paxillin, parvins, p130Cas, Src-family kinases and GTPases of the Rho family. Several integrin subunits like alpha 1, alpha 5 , beta 1 and beta 3 have been directly implicated in the cardiac pathophysiology, however, the underlying molecular mechanism and downstream signaling cascades are poorly understood (22- 25) (Table 1). In the cardiovascular system, integrins are expressed in cardiac myocytes and fibroblasts as well as cells composing the vasculature, blood and neurons. The importance of integrin regulation and function in cardiac cells is given in the following sections. Integrin function is required for proper cardiac development and myocyte attachment to extracellular matrix, growth and viability (26). Integrin-dependent pathways also mediate hypertrophic responses to mechanical stimuli associated with cardiac myocyte strain (10, 11) and are required for stimulation of hypertrophy by phenylephrine (PE) or endothelin-1 (ET-1) (27-29). Cardiac myocytes express integrins alpha 1 , alpha 3 , alpha 5 , alpha 6 , alpha 7 , alpha 9 , alpha 10 , beta 1 , beta 3 , and beta 5 (7), many of which are regulated by developmental and pathological stimuli (30, 31). Adult myocytes express the laminin binding alpha 7 beta 1 heterodimer as the major integrin, while the alpha 5 beta 1 fibronectin receptor and the alpha 6 beta 1 laminin receptor are expressed in cardiac myocytes during embryonic development (32, 33). The primary beta integrin subunit found in myocytes is beta 1 . Different splice variants are expressed in the embryonic (beta 1A ) and adult myocytes (beta 1D ) (34), which differ in specific amino acid sequences at the cytoplasmic domain and their interaction with cytoskeletal and signaling molecules (35). The interaction of cardiac fibroblasts with the surrounding matrix is critical for repair mechanisms, including synthesis of matrix proteins, proliferation, collagen gel contraction and cell motility (36, 37). Cardiac fibroblast activation in the failing heart is associated with increased expression of extracellular matrix proteins (38- 41). Cardiac fibroblasts express integrins alpha , alpha , alpha 3 , alpha 5 , alpha 8 , alpha 10 , beta 1 , beta 3 and beta 5 (37, 42-45). Angiotensin II (Ang II) and other growth factors stimulate cardiac fibroblast contraction and adhesion via beta 1 and alpha v beta 3 integrins, which involve inside-to- outside signaling mechanisms (37, 43-45). The term “integrin” was coined to reflect the capacity of a receptor family to integrate the extracellular and intracellular environment by bidirectional signaling (Hynes, 1987 #496). Interactions with extracellular matrix (ECM), cytoskeletal and various signal transduction cascades enables integrins to mediate both outside in and inside-out signaling (2, 7, 46). Inside-out signaling occurs when specific intracellular signals impinge on integrin cytoplasmic domains, triggering changes in conformation and ligand-binding affinity in the extracellular domain. For example Ang II induces a significant increase in β 1 integrin-dependent adhesion of cardiac fibroblasts to collagen I (37), by inducing a high-affinity state in the integrin molecule. In turn, binding of extracellular ligands produces intracellular signals (ie, outside-in signals) such as changes in intracellular signaling events and cytoskeletal reorganization that critically influence cell shape, migration, growth, and survival (Hynes, 2002 #1). There is number of excellent full reviews dedicated to basic mechanism of integrin bidirectional signaling (2, 47, 48). The specificity of integrin signaling is made possible by alpha and beta subunits that form the heterodimeric pair. Analysis of the amino acid sequences of cytoplasmic tails reveals considerable diversity among integrin subunits (49), suggesting differences in signal transduction among these ECM receptors. Because integrins lack enzymatic activity, activation of signal factors requires interaction with cellular proteins that have kinase activity. The cytoplasmic tail of the beta subunit directly binds to several cytoskeletal proteins that associate with signaling molecules (50). Integrins signal through a wide array of intracellular second messenger systems including calcium channels, phosphatidylinositol-4,5-bisphosphate, phospholipase-C (PLC), the Na/H antiporter, tyrosine and serine/threonine kinases, phosphatases, Rho GTPases, mitogen-activated protein (MAP) kinases, and cyclin D1 (13, 51-57). In blood vessels and cardiac cells, shear stress and stretch are important activators of integrins and signal transduction pathways. Mechanical load applied to integrin ligands (ECM) triggers the assembly and growth of focal contacts (58, 59) and activation of FAK and MAP kinases (18, 60). Although integrins works as “receptors” inducing multiple biological functions, the transduction processes are poorly understood. Stretch-induced conformational changes in the ECM may alter integrin structure, resulting in activation of liganded integrin receptors and focal contact-associated secondary messenger pathways in the cell, such as FAK, Src family kinases, Abl and ILK (50, 61). However, other mechanisms may be operational. For example, membrane-bound proteins such as the ADAMs (a disintegrin and a metalloproteinase domain) also support integrin-mediated cell adhesion since these molecules can cleave ECM proteins by their metalloproteinase domains (62). In addition, mechanical stimulation increases growth factor shedding into the ECM (63). Because ADAMs have the ability to shed many cell-adhesion molecules and cell- surface proteins including cytokines and growth factors, signaling pathways activated by these factors could interact with those of integrins. Integrin signaling may also crosstalk with signals generated by stretch-induced secretion of autocrine factors. Uniaxial stretch stimulates autocrine release of Ang II and ET-1, which induce cellular hypertrophy via phosphorylation cascades (64, 65). Interestingly, AT 1 mediates cardiac hypertrophy by upregulation of beta 1 integrin expression (66) and acts as a mechanoreceptor in cardiac tissue (67). A considerable amount of integrin signaling involves lipid domains located on the cell surface. These microenvironments function as signal organizing centers and platforms by exploiting multiple protein–lipid and protein–protein interactions to link the cytoplasmic tail of transmembrane receptors with other protein scaffolds. These interactions serve to assemble kinases, phosphatases, and other catalytically active molecules in order to generate specific signals that are temporally and spatially controlled (68-70). Integrins signal through at least two types ...

Similar publications

Article
The mammalian central nervous system (CNS) is comprised of billions of neurons and glia that are intertwined with an elaborate network of blood vessels. These various neural and vascular cell types actively converse with one another to form integrated, multifunctional complexes, termed neurovascular units. Cell-cell communication within neurovascul...
Article
Full-text available
Cancer cells interact with endothelial cells during the process of metastatic spreading. Here, we use a small interfering RNA screen targeting Rho GTPases in cancer cells to identify Cdc42 as a critical regulator of cancer cell-endothelial cell interactions and transendothelial migration. We find that Cdc42 regulates β1 integrin expression at the t...

Citations

... Unexpectedly, none of these receptors were differential modulated by miR-223 gain-and loss-offunction assays, supporting the notion that they do not play a role in miR-223 mediated modulation of PE/ST cell migration. We subsequently tested whether such modulation might be mediated by cellextracellular matrix receptors, i.e. integrins, given their pivotal role in cell migration in distinct cardiovascular contexts [74][75][76]. Similarly, Itga1, Itga4 and Itgb1 were not differential modulated by miR-223 gain-and loss-of-function assays. ...
Preprint
Full-text available
Cardiac development is a complex developmental process that results in the formation of the four-chambered organ from a single linear heart tube. In the early stages of development, the linear heart tube is composed of only two tissue layers, an external myocardium that is internally lined by the endocardium. Subsequently, the proepicardium emerges at the septum transversum and soon thereafter proepicardial cells will migrate into the naked myocardium, leading to the formation of the embryonic epicardium. As cardiac development proceeds, epicardial-derived cells migrate into the subepicardial space and subsequently invade the developing ventricular chambers leading to the contribution of distinct cardiovascular cell types such as the cardiac fibroskeleton and different components of coronary vasculature. At present, the molecular mechanisms that regulate the transitional process from the proepicardium to the embryonic myocardium are largely unexplored. In this study we have implemented an ex vivo proepicardium/septum transversum (PE/ST)-embryonic myocardium explant model and we demonstrated that miR-223, but not miR-195, is capable of modulating PE/ST migration, a process that seems to be mediated by Slug expression. Thus, our study demonstrates for the first time the implication of distinct microRNAs in the PE/ST to embryonic myocardium transition in chicken embryonic hearts.
... Recently, Campillo et al. have demonstrated that, under IS and PC stimulation, monocyte cells increase podosome formation implicated in cell migration and invasion mediated by activation of integrin-linked kinase (ILK)/AKT signaling [17]. Also, activated ILK stimulates several signaling pathways including NF-κB [43]. Other studies have reported that the stimulation of monocytes with microparticles isolated from the plasma of patients with systemic lupus erythematosus favors the cell migration to peripheral tissues and perpetuate the inflammatory state mediated by the NF-κB pathway [44]. ...
Article
Full-text available
Atherosclerosis is initiated by the activation of endothelial cells that allows monocyte adhesion and transmigration through the vascular wall. The accumulation of uremic toxins such as indoxyl sulphate (IS) and p-cresol (PC) has been associated with atherosclerosis. Currently, miRNAs play a crucial role in the regulation of monocyte activation, adhesion, and trans-endothelial migration. The aim of the present study is to evaluate the effect of IS and PC on monocyte adhesion and migration processes in monocytes co-cultured with endothelial cells as well as to determine the underlying mechanisms. The incubation of HUVECs and THP-1 cells with both IS and PC toxins resulted in an increased migratory capacity of THP-1 cells. Furthermore, the exposure of THP-1 cells to both uremic toxins resulted in the upregulation of BMP-2 and miRNAs-126-3p, -146b-5p, and -223-3p, as well as the activation of nuclear factor kappa B (NF-κB) and a decrease in its inhibitor IĸB. Uremic toxins, such as IS and PC, enhance the migratory and adhesion capacity of THP-1 cells to the vascular endothelium. These toxins, particularly PC, contribute significantly to uremia-associated vascular disease by increasing in THP-1 cells the expression of BMP-2, NF-κB, and key miRNAs associated with the development of atherosclerotic vascular diseases.
... Integrin Linked Kinase (ILK) is a serine/threonine kinase that binds to the cytoplasmic domain of β-integrins and participates in cardiac remodelling after MI [30,56]. ILK lies upstream of many intracellular signalling pathways potentially involved in such effect, including angiogenesis, mechanotransduction, extracellular matrix-mediated signalling, cell survival, proliferation, differentiation and apoptosis [26,29]. The ILK role in cardiomyocyte contractile function is well studied. ...
Article
Full-text available
Endothelial dysfunction is an early event in coronary microvascular disease. Integrin-linked kinase (ILK) prevents endothelial nitric oxide synthase (eNOS) uncoupling and, thus, endothelial dysfunction. However, the specific role of endothelial ILK in cardiac function remains to be fully elucidated. We hypothesised that endothelial ILK plays a crucial role in maintaining coronary microvascular function and contractile performance in the heart. We generated an endothelial cell-specific ILK conditional knock-out mouse (ecILK cKO) and investigated cardiovascular function. Coronary endothelial ILK deletion significantly impaired cardiac function: ejection fraction, fractional shortening and cardiac output decreased, whilst left ventricle diastolic internal diameter decreased and E/A and E/Eʹ ratios increased, indicating not only systolic but also diastolic dysfunction. The functional data correlated with extensive extracellular matrix remodelling and perivascular fibrosis, indicative of adverse cardiac remodelling. Mice with endothelial ILK deletion suffered early ischaemic-like events with ST elevation and transient increases in cardiac troponins, which correlated with fibrotic remodelling. In addition, ecILK cKO mice exhibited many features of coronary microvascular disease: reduced cardiac perfusion, impaired coronary flow reserve and arterial remodelling with patent epicardial coronary arteries. Moreover, endothelial ILK deletion induced a moderate increase in blood pressure, but the antihypertensive drug Losartan did not affect microvascular remodelling whilst only partially ameliorated fibrotic remodelling. The plasma miRNA profile reveals endothelial-to-mesenchymal transition (endMT) as an upregulated pathway in endothelial ILK conditional KO mice. Our results show that endothelial cells in the microvasculature in endothelial ILK conditional KO mice underwent endMT. Moreover, endothelial cells isolated from these mice and ILK-silenced human microvascular endothelial cells underwent endMT, indicating that decreased endothelial ILK contributes directly to this endothelial phenotype shift. Our results identify ILK as a crucial regulator of microvascular endothelial homeostasis. Endothelial ILK prevents microvascular dysfunction and cardiac remodelling, contributing to the maintenance of the endothelial cell phenotype.
... The decrease in adhesion to collagens is most likely due to changes in the α2 and β1 integrin levels; the α2β1 integrin is a default collagen-binding integrin [80]. Furthermore, the similarity between the two aforementioned cell lines is reflected in data for the fibronectin that is dependent on α5β1 integrin [81], the differences for which are coherent in α5 and β1, as well as combined α5β1. However, the same change in adhesion to laminin (following WWOX overexpression) might be due to the various α integrins in U251MG and U87MG. ...
Article
Full-text available
Following the discovery of WWOX, research has moved in many directions, including the role of this putative tumor suppressor in the central nervous system and related diseases. The task of determining the nature of WWOX in glioblastoma (GBM) is still considered to be at the initial stage; however, the influence of this gene on the GBM malignant phenotype has already been reported. Because most of the available in vitro research does not consider several cellular GBM models or a wide range of investigated biological assays, the present study aimed to determine the main processes by which WWOX exhibits anticancer properties in GBM, while taking into account the phenotypic heterogeneity between cell lines. Ectopic WWOX overexpression was studied in T98G, DBTRG-05MG, U251MG, and U87MG cell lines that were compared with the use of assays investigating cell viability, proliferation, apoptosis, adhesion, clonogenicity, three-dimensional and anchorage-independent growth, and invasiveness. Observations presenting the antineoplastic properties of WWOX were consistent for T98G, U251MG, and U87MG. Increased proliferation and tumor growth were noted in WWOX-overexpressing DBTRG-05MG cells. A possible explanation for this, arrived at via bioinformatics tools, was linked to the TARDBP transcription factor and expression differences of USP25 and CPNE2 that regulate EGFR surface abundance. Collectively, and despite various cell line-specific circumstances, WWOX exhibits its anticancer nature mainly via a reduction of cell viability and invasiveness of glioblastoma.
... Integrin β1 senses ECM stiffness by recognising collagen accumulation in fibrotic tissue and triggers intracellular signalling that promotes fibrosis 10,11,35,36 . Treatment with the integrin β1 inhibitor BTT-3033 37 significantly reduced VGLL3 nuclear translocation, indicating the involvement of integrin β1 in this process (Fig. 1n, Supplementary Fig. 2l). ...
Article
Full-text available
Myofibroblasts cause tissue fibrosis by producing extracellular matrix proteins, such as collagens. Humoral factors like TGF-β, and matrix stiffness are important for collagen production by myofibroblasts. However, the molecular mechanisms regulating their ability to produce collagen remain poorly characterised. Here, we show that vestigial-like family member 3 (VGLL3) is specifically expressed in myofibroblasts from mouse and human fibrotic hearts and promotes collagen production. Further, substrate stiffness triggers VGLL3 translocation into the nucleus through the integrin β1-Rho-actin pathway. In the nucleus, VGLL3 undergoes liquid-liquid phase separation via its low-complexity domain and is incorporated into non-paraspeckle NONO condensates containing EWS RNA-binding protein 1 (EWSR1). VGLL3 binds EWSR1 and suppresses miR-29b, which targets collagen mRNA. Consistently, cardiac fibrosis after myocardial infarction is significantly attenuated in Vgll3-deficient mice, with increased miR-29b expression. Overall, our results reveal an unrecognised VGLL3-mediated pathway that controls myofibroblasts’ collagen production, representing a novel therapeutic target for tissue fibrosis.
... Pathological changes in the heart are determined by the improperly over-activated pro-survival genes in vivo cardiac myocytes and broblasts in the surviving myocardium around the infarcted lesion [2] . Cardiomyocyte-speci c knockout of pro-survival integrin β1 subunit and its downstream molecules such as focal adhesion kinase (FAK) have been found to exacerbate myocardial remodeling after MI via their aggravation of apoptosis in the cardiomyocytes [3][4][5]. Pro-survival adaptor protein CrkL (v-crk avian sarcoma virus CT-10 oncogene homolog (Crk)-like) can bind to C3G (rap guanine nucleotide exchange factor 1), which acts as a guanine nucleotide exchanger in integrin pathway [3]. Our previous study has shown that knockdown of CrkL can worsen cell apoptosis, which was induced by hypoxia / reoxygenation (H/R) in vitro cardiomyocytic cell line [6]. ...
... Cardiomyocyte-speci c knockout of pro-survival integrin β1 subunit and its downstream molecules such as focal adhesion kinase (FAK) have been found to exacerbate myocardial remodeling after MI via their aggravation of apoptosis in the cardiomyocytes [3][4][5]. Pro-survival adaptor protein CrkL (v-crk avian sarcoma virus CT-10 oncogene homolog (Crk)-like) can bind to C3G (rap guanine nucleotide exchange factor 1), which acts as a guanine nucleotide exchanger in integrin pathway [3]. Our previous study has shown that knockdown of CrkL can worsen cell apoptosis, which was induced by hypoxia / reoxygenation (H/R) in vitro cardiomyocytic cell line [6]. ...
... Integrin pathway links extracellular matrix substances to cytoskeleton in cells, and senses mechanical stretch, and translates it into intracellular biochemical signaling [3,16]. Cardiomyocyte-speci c deletion of pro-survival integrin β1 subunit and its downstream molecules such as FAK may increase apoptosis in vivo cardiomyocytes and aggravate cardiomyopathy induced by MI, isoproterenol or pressure overload [4,5]. ...
Preprint
Full-text available
Cardiomyocyte-specific knockout of pro-survival integrin β1 subunit and its downstream components have been demonstrated to aggravate remodeling after myocardial infarction (MI). However, as a component of integrin pathway, it is unclear whether knockdown of pro-survival C3G (rap guanine nucleotide exchange factor 1) in cardiac myocytes and fibroblasts could have effect on myocardial remodeling. A rat model of MI was established by ligation of left anterior descending coronary artery. Infarcted myocardium and its border zones in Sprague-Dawley rats were transiently infected with C3G knockout lentivirus via local injection to knockdown C3G in the myocardium. Twelve weeks after injection with the lentiviruses, cardiomyocytic apoptosis and collagen in surviving myocardium, and left ventricle (LV) end-diastolic diameter were decreased, whereas LV weight / body weight ratio and LV ejection fraction were increased in MI group via down-regulation of pro-survival C3G, phosphorylated (p) ERK1/2 (phosphorylated extracellular regulated kinase 1/2) and Bcl-2 (B-cell lymphoma-2), and up-regulation of pro-apoptotic Bax in the surviving myocardium. On the other hand, treatment with the lentiviruses was found to delete C3G and diminish cell proliferation in vitro cardiac myocytic and fibroblastic cell lines respectively via down-regulation of p-ERK1/2 and Bcl-2, and up-regulation of Bax. In conlusion, knockdown of pro-survival C3G in myocardium may mitigate surviving myocardial remodeling after MI, possibly through regulation of p-ERK1/2, Bcl-2 and Bax in vivo cardiac myocytes and fibroblasts.
... Pathological changes in the heart are determined by the improperly over-activated pro-survival genes in vivo cardiac myocytes and broblasts in the surviving myocardium around the infarcted lesion [2]. Cardiomyocyte-speci c knockout of pro-survival integrin β1 subunit and its downstream molecules such as focal adhesion kinase (FAK) have been found to exacerbate myocardial remodeling after MI via their aggravation of apoptosis in the cardiomyocytes [3][4][5]. Pro-survival adaptor protein CrkL (v-crk avian sarcoma virus CT-10 oncogene homolog (Crk)-like) can bind to C3G (rap guanine nucleotide exchange factor 1), which acts as a guanine nucleotide exchanger in integrin pathway [3]. Our previous study has shown that knockdown of CrkL can worsen cell apoptosis, which was induced by hypoxia / reoxygenation (H/R) in vitro cardiomyocytic cell line [6]. ...
... Cardiomyocyte-speci c knockout of pro-survival integrin β1 subunit and its downstream molecules such as focal adhesion kinase (FAK) have been found to exacerbate myocardial remodeling after MI via their aggravation of apoptosis in the cardiomyocytes [3][4][5]. Pro-survival adaptor protein CrkL (v-crk avian sarcoma virus CT-10 oncogene homolog (Crk)-like) can bind to C3G (rap guanine nucleotide exchange factor 1), which acts as a guanine nucleotide exchanger in integrin pathway [3]. Our previous study has shown that knockdown of CrkL can worsen cell apoptosis, which was induced by hypoxia / reoxygenation (H/R) in vitro cardiomyocytic cell line [6]. ...
... Integrin pathway links extracellular matrix substances to cytoskeleton in cells, and senses mechanical stretch, and translates it into intracellular biochemical signaling [3,16]. Cardiomyocyte-speci c deletion of pro-survival integrin β1 subunit and its downstream molecules such as FAK may increase apoptosis in vivo cardiomyocytes and aggravate cardiomyopathy induced by MI, isoproterenol or pressure overload [4,5]. ...
Preprint
Full-text available
Background: Cardiomyocyte-specific knockout of pro-survival integrin β1 subunit and its downstream components have been demonstrated to aggravate remodeling after myocardial infarction (MI). However, as a component of integrin pathway, it is unclear whether knockdown of pro-survival C3G (rap guanine nucleotide exchange factor 1) in cardiac myocytes and fibroblasts could have effect on myocardial remodeling. Methods and results: A rat model of MI was established by ligation of left anterior descending coronary artery. Infarcted myocardium and its border zones in Sprague-Dawley rats were transiently infected with C3G knockout lentivirus via local injection to knockdown C3G in the myocardium. Twelve weeks after injection with the lentiviruses, cardiomyocytic apoptosis and collagen in surviving myocardium, and left ventricle (LV) end-diastolic diameter were decreased, whereas LV weight / body weight ratio and LV ejection fraction were increased in MI group via down-regulation of pro-survival C3G, phosphorylated (p) ERK1/2 (phosphorylated extracellular regulated kinase 1/2) and Bcl-2 (B-cell lymphoma-2), and up-regulation of pro-apoptotic Bax in the surviving myocardium. On the other hand, treatment with the lentiviruses was found to delete C3G and diminish cell proliferation in vitro cardiac myocytic and fibroblastic cell lines respectively via down-regulation of p-ERK1/2 and Bcl-2, and up-regulation of Bax. Conclusions: Knockdown of pro-survival C3G in myocardium may mitigate surviving myocardial remodeling after MI, possibly through regulation of p-ERK1/2, Bcl-2 and Bax in vivo cardiac myocytes and fibroblasts.
... In addition, Nck2 is involved in linking AXL with other signaling complexes. The interaction of AXL and Nck2 can connect AXL to a ternary complex consisting of the particularly interesting new cysteine-histidine-rich protein (PINCH) and integrin-linked kinase (ILK), which is a major component of signaling platforms at focal adhesions, thereby enabling AXL to regulate cytoskeleton dynamics (McCarty, 1998;Lal et al., 2009;Verma et al., 2011). In inflammatory breast cancer cell lines, depletion of AXL-stabilizing protein TIG1 reduces the expression of Matrix metalloproteinase 9 (MMP9), which has been identified as an essential regulator for the AXLmediated invasion (Tai et al., 2008;Koorstra et al., 2009;Han et al., 2013;Wang et al., 2013). ...
Article
Full-text available
Flaviviruses, as critically important pathogens, are still major public health problems all over the world. For instance, the evolution of ZIKV led to large-scale outbreaks in the Yap island in 2007. DENV was considered by the World Health Organization (WHO) as one of the 10 threats to global health in 2019. Enveloped viruses hijack a variety of host factors to complete its replication cycle. Phosphatidylserine (PS) receptor, AXL, is considered to be a candidate receptor for flavivirus invasion. In this review, we discuss the molecular structure of ZIKV and DENV, and how they interact with AXL to successfully invade host cells. A more comprehensive understanding of the molecular mechanisms of flavivirus-AXL interaction will provide crucial insights into the virus infection process and the development of anti-flavivirus therapeutics.
... It is known that integrin signalling triggers several pathways, including the PLC/PKC/c-Src pathway, [35][36][37] Figure S2). Therefore, this evidence indicates that PLC, PKC and c-Src signal pathways are responsible for migratory potential after TSP-2 treatment. ...
Article
Full-text available
Osteosarcoma is an extremely common primary bone malignancy that is highly metastatic, with most deaths resulting from pulmonary metastases. The extracellular matrix protein thrombospondin‐2 (TSP‐2) is key to many biological processes, such as inflammation, wound repair and tissue remodelling. However, it is unclear as to what biological role TSP‐2 plays in human metastatic osteosarcoma. The immunochemistry analysis from osteosarcoma specimens identified marked up‐regulation of TSP‐2 in late‐stage osteosarcoma. Furthermore, we found that TSP‐2 increased the levels of matrix metallopeptidase 9 (MMP‐9) expression and thereby increased the migratory potential of human osteosarcoma cells. Osteosarcoma cells pre‐treated with an MMP‐9 monoclonal antibody (mAb), an MMP‐9 inhibitor, or transfected with MMP‐9 small interfering RNA (siRNA) reduced the capacity of TSP‐2 to potentiate cell migration. TSP‐2 treatment activated the PLCβ, PKCα, c‐Src and nuclear kappa factor B (NF‐κB) signalling pathways, while the specific siRNA, inhibitors and mutants of these cascades reduced TSP‐2‐induced stimulation of migration activity. Knockdown of TSP‐2 expression markedly reduced cell metastasis in cellular and animal experiments. It appears that an interaction between TSP‐2 and integrin αvβ3 activates the PLCβ, PKCα and c‐Src signalling pathways and subsequently activates NF‐κB signalling, increasing MMP‐9 expression and stimulating migratory activity amongst human osteosarcoma cells.
... It forms the IPAP1 complex with PINCH1 and α-parvin proteins, which are linked to cell adhesion, motility, and fibroblast and epithelial cell survival . Both widely conserved and distributed throughout mammalian tissues, ILK expression is greatest in the heart and cardiac-specific ablation in mice causes cardiomyopathy and sudden death (Hannigan, Coles, & Dedhar, 2007;Lal et al., 2009). ILK, which is understood to mediate hypertrophic responses to mechanical stress, is thought to protect the heart by promoting cell survival via activation of AKT, a key regulator of oxidative stress and myocardial hypertrophy (Bettink et al., 2010;Lal et al., 2009;Sopko et al., 2011). ...
... Both widely conserved and distributed throughout mammalian tissues, ILK expression is greatest in the heart and cardiac-specific ablation in mice causes cardiomyopathy and sudden death (Hannigan, Coles, & Dedhar, 2007;Lal et al., 2009). ILK, which is understood to mediate hypertrophic responses to mechanical stress, is thought to protect the heart by promoting cell survival via activation of AKT, a key regulator of oxidative stress and myocardial hypertrophy (Bettink et al., 2010;Lal et al., 2009;Sopko et al., 2011). ILK protein levels have been shown to be elevated in hypertrophic ventricles of patients with congenital and acquired outflow tract obstruction (Lu et al., 2006), patient dilated cardiomyopathy heart samples (Chadin, Belokurova, Stepanova, Ivanova, & Shirinskii, 2006;Sopko et al., 2011), and mouse models of myocardial infarction (left anterior descending artery ligation) or pressure overload (transaortic constriction) (Sopko et al., 2011). ...
Article
Full-text available
Heart disease remains the number one killer of women in the US. Nonetheless, studies in women and female animal models continue to be underrepresented in cardiac research. Hypertrophic cardiomyopathy (HCM), the most commonly inherited cardiac disorder, has been tied to sarcomeric protein variants in both sexes. Among the susceptible genes, TNNC1—encoding cardiac troponin C (cTnC)—causes a substantial HCM phenotype in mice. Mice bearing an HCM‐associated cTnC‐A8V point mutation exhibited a significant decrease in stroke volume and left ventricular diameter and volume. Importantly, isovolumetric contraction time was significantly higher for female HCM mice. We utilized a transcriptomic approach to investigate the basis underlying the sexual dimorphism observed in the cardiac physiology of adult male and female HCM mice. RNA sequencing revealed several altered canonical pathways within the HCM mice versus WT groups including an increase in eukaryotic initiation factor 2 signaling, integrin‐linked kinase signaling, actin nucleation by actin‐related protein‐Wiskott‐Aldrich syndrome family protein complex, regulation of actin‐based motility by Rho kinase, vitamin D receptor/retinoid X receptor activation, and glutathione redox reaction pathways. In contrast, valine degradation, tricarboxylic acid cycle II, methionine degradation, and inositol phosphate compound pathways were notably down‐regulated in HCM mice. These down‐regulated pathways may be reduced in response to altered energetics in the hypertrophied hearts and may represent conservation of energy as the heart is compensating to meet increased contractile demands. HCM male versus female mice followed similar trends of the canonical pathways altered between HCM and WT. In addition, seven of the differentially expressed genes in both WT and HCM male versus female comparisons swapped directions in fold‐change between the sexes. These findings suggest a sexually‐dimorphic HCM phenotype due to a sarcomeric mutation and pinpoint several key targetable pathways and genes that may provide the means to alleviate the more severe decline in female cardiac function. Sex‐specific patterns in transcriptome regulation.