Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
1.
Curr Opin Cell Biol ; 85: 102273, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37918273

RESUMO

Recent data indicate that integrin and non-integrin collagen receptors cooperate in the fibrosis-specific microenvironment (i.e., the fibrotic niche). In certain tumor types, DDR1 can regulate the interaction with collagen III to regulate dormancy and metastasis, whereas in other tumor types, DDR1 can be shed and used to reorganize collagen. DDR1 expressed on tumor cells, together with DDR2 and α11ß1 integrin expressed on cancer-associated fibroblasts, can increase tumor tissue stiffness. Integrin α1ß1 and α2ß1 are present on immune cells where they together with the immunosuppressive collagen receptor LAIR-1 can mediate binding to intratumor collagens. In summary, collagen-binding integrins together with DDRs, can create fibrillar collagen niches that act as traps to hinder immune cell trafficking into the tumor cell mass. Binding of collagens via LAIR-1 on immune cells in turn results in CD8+T-cell exhaustion. Continued studies of these complex interactions are needed for successful new stroma-based therapeutic interventions. In the current review, we will summarize recent data on collagen receptors with a special focus on their potential role in tumor fibrosis and highlight their collaborative roles in tumor fibrotic niches.


Assuntos
Colágeno , Neoplasias , Humanos , Ligação Proteica , Colágeno/metabolismo , Receptores de Colágeno/metabolismo , Integrinas/metabolismo , Transdução de Sinais , Fibrose , Microambiente Tumoral
2.
J Thromb Haemost ; 21(12): 3619-3632, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37678551

RESUMO

BACKGROUND: Upon vessel injury, platelets adhere to exposed matrix constituents via specific membrane receptors, including the von Willebrand factor receptor glycoprotein (GP)Ib-IX-V complex and integrins ß1 and ß3. In platelets, the Fes/CIP4-homology Bin-Amphiphysin-Rvs protein PACSIN2 associates with the cytoskeletal and scaffolding protein filamin A (FlnA), linking GPIbα and integrins to the cytoskeleton. OBJECTIVES: Here we investigated the role of PACSIN2 in platelet function. METHODS: Platelet parameters were evaluated in mice lacking PACSIN2 and platelet integrin ß1. RESULTS: Pacsin2-/- mice displayed mild thrombocytopenia, prolonged bleeding time, and delayed thrombus formation in a ferric chloride-mediated carotid artery injury model, which was normalized by injection of control platelets. Pacsin2-/- platelets formed unstable thrombi that embolized abruptly in a laser-induced cremaster muscle injury model. Pacsin2-/- platelets had hyperactive integrin ß1, as evidenced by increased spreading onto surfaces coated with the collagen receptor α2ß1-specific peptide GFOGER and increased binding of the antibody 9EG7 directed against active integrin ß1. By contrast, Pacsin2-/- platelets had normal integrin αIIbß3 function and expressed P-selectin normally following stimulation through the collagen receptor GPVI or with thrombin. Deletion of platelet integrin ß1 in Pacsin2-/- mice normalized platelet count, hemostasis, and thrombus formation. A PACSIN2 peptide mimicking the FlnA-binding site mediated the pull-down of a FlnA rod 2 construct by integrin ß7, a model for integrin ß-subunits. CONCLUSIONS: Pacsin2-/- mice displayed severe thrombus formation defects due to hyperactive platelet integrin ß1. The data suggest that PACSIN2 binding to FlnA negatively regulates platelet integrin ß1 hemostatic function.


Assuntos
Integrina beta1 , Ativação Plaquetária , Trombose , Animais , Camundongos , Plaquetas/metabolismo , Hemostasia , Hemostáticos/metabolismo , Integrina beta1/metabolismo , Peptídeos/farmacologia , Adesividade Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores de Colágeno/metabolismo , Trombose/metabolismo
3.
Expert Rev Hematol ; 16(8): 593-605, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37335893

RESUMO

INTRODUCTION: Antithrombotic therapy field is undergoing rapid and significant changes during the past decade. In addition to new therapeutic strategies with existing targets, investigators are exploring the potential use of new targets to address unmet needs to treat patients with arterial diseases. AREAS COVERED: We aim to provide an update on and a comprehensive review of the antithrombic agents that are being explored in patients with arterial diseases. We discuss latest developments with respect to upstream antiplatelet agents, and collagen and thrombin pathway inhibitors. We searched PubMed databases for English language articles using keywords: antiplatelet agents, thrombin pathway inhibitors, collagen receptors, arterial disease. EXPERT OPINION: Despite implementation of potent P2Y12 inhibitors, there are numerous unmet needs in the treatment of arterial diseases including ceiling effect of currently available antiplatelet agents along with and an elevated risk of bleeding. The latter observations encouraged investigators to explore new targets that can attenuate the generation of platelet-fibrin clot formation and subsequent ischemic event occurrences with minimal effect on bleeding. These targets include collagen receptors on platelets and thrombin generation including FXa, FXIa, and FXIIa. In addition, investigators are studying novel antiplatelet agents/strategies to facilitate upstream therapy in high-risk patients.


Assuntos
Inibidores da Agregação Plaquetária , Trombose , Humanos , Inibidores da Agregação Plaquetária/efeitos adversos , Trombina/metabolismo , Trombina/farmacologia , Trombina/uso terapêutico , Trombose/tratamento farmacológico , Trombose/etiologia , Plaquetas/metabolismo , Hemorragia/etiologia , Hemorragia/tratamento farmacológico , Receptores de Colágeno/metabolismo
4.
Int J Mol Sci ; 23(19)2022 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-36233024

RESUMO

The tumor microenvironment, including cancer-associated fibroblast (CAF), plays an active role in non-small cell lung cancer (NSCLC) development and progression. We previously reported that collagen type XI and integrin α11, a collagen receptor, were upregulated in NSCLC; the latter promotes tumor growth and metastasis. We here explored the role of collagen type XI in NSCLC stroma. We showed that the presence of collagen type XI in collagen type I matrices inhibits CAF-mediated collagen remodeling and cell migration. This resulted in the inhibition of CAF-dependent lung-tumor cell invasion. Among the collagen receptors expressed on CAF, we determined that DDR2 and integrin α2ß1, but not integrin α11ß1, mediated the high-affinity binding to collagen type XI. We further demonstrated that collagen type XI restrained the integrin binding site availability on collagen type I matrices, thus limiting cell interaction with collagen type I. As a consequence, CAFs failed to activate FAK, p38 and Akt one hour after they interacted with collagen type I/XI. We concluded that collagen type XI may have a competitive negative feedback role on the binding of collagen type I to its receptors.


Assuntos
Fibroblastos Associados a Câncer , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Sítios de Ligação , Fibroblastos Associados a Câncer/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Colágeno Tipo XI/metabolismo , Humanos , Integrina alfa2beta1/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Colágeno/metabolismo , Microambiente Tumoral
5.
Thromb Res ; 218: 112-129, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36037547

RESUMO

The collagen receptor glycoprotein VI (GPVI) drives strong platelet activation, however its role at later stages of clotting remains less clear. Controlled timing of addition of anti-human GPVI Fab (clone E12) with microfluidic venous whole blood flow over collagen (± lipidated tissue factor, TF) produced distinct effects on platelets, fibrin, P-selectin exposure, and phosphatidylserine (PS) exposure. On collagen alone, Fab present initially potently reduced platelet deposition on collagen, while Fab added 90 s after initial platelet deposition, stopped subsequent platelet accumulation (despite the absence of fibrin). With thrombin generation via TF, Fab added at either t = 0 or 90 s had no effect on platelet deposition. However, Fab added initially, but not at 90-s, blocked fibrin formation. Gly-Pro-Arg-Pro ablated fibrin formation without effect on platelet accumulation (regardless of Fab added at t = 0 or 90 s), indicating thrombin signaling can suffice over GPVI signaling. Still, Fab moderately reduced P-selectin exposure with thrombin present and fibrin absent. On collagen/TF, Fab present initially ablated PS exposure, but had no effect when added 30 to 90-s later. The thrombin generated via PS exposure had an important role in driving platelet deposition in the presence of Fab, since inhibition of PS via annexin V binding in the presence of Fab significantly inhibited platelet deposition. We conclude GPVI signaling in the first platelet layer on collagen dictates thrombin and fibrin production, but the role of GPVI at subsequent times after formation of the first monolayer is obscured by thrombin-induced signaling.


Assuntos
Trombina , Tromboplastina , Anexina A5 , Colágeno/metabolismo , Colágeno/farmacologia , Fibrina/metabolismo , Humanos , Microfluídica , Selectina-P/metabolismo , Fosfatidilserinas , Glicoproteínas da Membrana de Plaquetas , Receptores de Colágeno/metabolismo , Trombina/metabolismo , Tromboplastina/metabolismo
6.
Photodermatol Photoimmunol Photomed ; 38(2): 150-157, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34469613

RESUMO

BACKGROUND: Endo180 is involved in collagen remodeling by incorporating extracellular degraded collagen. Ultraviolet irradiation of dermal fibroblasts reduces Endo180 expression, which affects collagen fiber remodeling. However, it is unclear whether the decrease in Endo180 is directly related to the decrease in type I collagen fibers during photoaging. We aimed to clarify the relationship between Endo180 reduction and the decrease in type I collagen fibers observed in photoaged dermis. METHODS: Endo180 was reduced in normal human dermal fibroblasts using RNAi. Endo180 knockdown cells were inoculated into collagen gels. The influence of Endo180 knockdown was evaluated by measuring mRNA expression of collagen fiber remodeling-related factors and collagen gel contraction. The collagen state and oxidative stress in the collagen gels were also measured. RESULTS: Endo180 knockdown cells, which were confirmed by gelatin uptake inhibition, showed upregulation of matrix metalloproteinase-1 and downregulation of type I collagen mRNA expression when cultured in collagen gels. The contractility of the collagen gel was reduced by Endo180 knockdown. The collagen state in the extracellular matrix of the collagen gels containing Endo180 knockdown fibroblasts showed increased amounts of 3/4 fragmented collagen and denatured collagen and decreased type I collagen synthesis. In addition, an increase in intracellular oxidative stress was observed. CONCLUSIONS: This study confirmed that the decrease in Endo180 caused a failure in collagen fiber formation and a decrease in collagen production, reproducing the photoaging dermal structural changes. This suggests that the decrease in Endo180 may be involved in wrinkle formation, which is a characteristic of photoaged skin.


Assuntos
Colágeno Tipo I , Metaloproteinase 1 da Matriz , Receptores de Colágeno , Receptores Mitogênicos , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Fibroblastos/metabolismo , Humanos , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 1 da Matriz/metabolismo , Receptores de Colágeno/metabolismo , Receptores Mitogênicos/metabolismo
7.
Int J Mol Sci ; 22(21)2021 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-34768883

RESUMO

Malignant mesothelioma (MM) is a highly aggressive cancer with limited therapeutic options. We have previously shown that the endocytic collagen receptor, uPARAP, is upregulated in certain cancers and can be therapeutically targeted. Public RNA expression data display uPARAP overexpression in MM. Thus, to evaluate its potential use in diagnostics and therapy, we quantified uPARAP expression by immunohistochemical H-score in formalin-fixed paraffin-embedded bioptic/surgical human tissue samples and tissue microarrays. We detected pronounced upregulation of uPARAP in the three main MM subtypes compared to non-malignant reactive mesothelial proliferations, with higher expression in sarcomatoid and biphasic than in epithelioid MM. The upregulation appeared to be independent of patients' asbestos exposure and unaffected after chemotherapy. Using immunoblotting, we demonstrated high expression of uPARAP in MM cell lines and no expression in a non-malignant mesothelial cell line. Moreover, we showed the specific internalization of an anti-uPARAP monoclonal antibody by the MM cell lines using flow cytometry-based assays and confocal microscopy. Finally, we demonstrated the sensitivity of these cells towards sub-nanomolar concentrations of an antibody-drug conjugate formed with the uPARAP-directed antibody and a potent cytotoxin that led to efficient, uPARAP-specific eradication of the MM cells. Further studies on patient cohorts and functional preclinical models will fully reveal whether uPARAP could be exploited in diagnostics and therapeutic targeting of MM.


Assuntos
Lectinas de Ligação a Manose/metabolismo , Glicoproteínas de Membrana/metabolismo , Mesotelioma Maligno/metabolismo , Receptores de Superfície Celular/metabolismo , Adulto , Idoso , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Feminino , Expressão Gênica , Humanos , Imunoconjugados/metabolismo , Masculino , Lectinas de Ligação a Manose/fisiologia , Glicoproteínas de Membrana/fisiologia , Mesotelioma Maligno/diagnóstico , Mesotelioma Maligno/fisiopatologia , Pessoa de Meia-Idade , Receptores de Superfície Celular/fisiologia , Receptores de Colágeno/genética , Receptores de Colágeno/metabolismo , Receptores de Colágeno/fisiologia , Receptores Mitogênicos/genética , Transcriptoma , Regulação para Cima
8.
Sci Rep ; 11(1): 11965, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34099810

RESUMO

Colchicine inhibits coronary and cerebrovascular events in patients with coronary artery disease (CAD), and although known to have anti-inflammatory properties, its mechanisms of action are incompletely understood. In this study, we investigated the effects of colchicine on platelet activation with a particular focus on its effects on activation via the collagen glycoprotein (GP)VI receptor, P2Y12 receptor, and procoagulant platelet formation. Therapeutic concentrations of colchicine in vitro (equivalent to plasma levels) significantly decreased platelet aggregation in whole blood and in platelet rich plasma in response to collagen (multiplate aggregometry) and reduced reactive oxygen species (ROS) generation (H2DCF-DA, flow cytometry) in response to GPVI stimulation with collagen related peptide-XL (CRP-XL, GPVI specific agonist). Other platelet activation pathways including P-selectin expression, GPIIb/IIIa conformational change and procoagulant platelet formation (GSAO+/CD62P+) (flow cytometry) were inhibited with higher concentrations of colchicine known to inhibit microtubule depolymerization. Pathway specific mechanisms of action of colchicine on platelets, including modulation of the GPVI receptor pathway at low concentrations, may contribute to its protective role in CAD.


Assuntos
Colchicina/química , Doença da Artéria Coronariana/tratamento farmacológico , Glicoproteínas da Membrana de Plaquetas/metabolismo , Espécies Reativas de Oxigênio/química , Plaquetas/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Colchicina/metabolismo , Colchicina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Selectina-P/metabolismo , Peptídeos/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Receptores de Colágeno/genética , Receptores de Colágeno/metabolismo , Transdução de Sinais
9.
Cell Tissue Res ; 383(3): 1135-1153, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33306155

RESUMO

Collagen XIII is a conserved transmembrane collagen mainly expressed in mesenchymal tissues. Previously, we have shown that collagen XIII modulates tissue development and homeostasis. Integrins are a family of receptors that mediate signals from the environment into the cells and vice versa. Integrin α11ß1 is a collagen receptor known to recognize the GFOGER (O=hydroxyproline) sequence in collagens. Interestingly, collagen XIII and integrin α11ß1 both have a role in the regulation of bone homeostasis. To study whether α11ß1 is a receptor for collagen XIII, we utilized C2C12 cells transfected to express α11ß1 as their only collagen receptor. The interaction between collagen XIII and integrin α11ß1 was also confirmed by surface plasmon resonance and pull-down assays. We discovered that integrin α11ß1 mediates cell adhesion to two collagenous motifs, namely GPKGER and GF(S)QGEK, that were shown to act as the recognition sites for the integrin α11-I domain. Furthermore, we studied the in vivo significance of the α11ß1-collagen XIII interaction by crossbreeding α11 null mice (Itga11-/-) with mice overexpressing Col13a1 (Col13a1oe). When we evaluated the bone morphology by microcomputed tomography, Col13a1oe mice had a drastic bone overgrowth followed by severe osteoporosis, whereas the double mutant mouse line showed a much milder bone phenotype. To conclude, our data identifies integrin α11ß1 as a new collagen XIII receptor and demonstrates that this ligand-receptor pair has a role in the maintenance of bone homeostasis.


Assuntos
Osso e Ossos , Colágeno Tipo XIII/metabolismo , Cadeias alfa de Integrinas/metabolismo , Integrinas/metabolismo , Receptores de Colágeno/metabolismo , Animais , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Adesão Celular , Linhagem Celular , Humanos , Camundongos , Camundongos Knockout
10.
Sci Signal ; 13(643)2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32753479

RESUMO

Cerebral amyloid angiopathy (CAA) and ß-amyloid (Aß) deposition in the brain parenchyma are hallmarks of Alzheimer's disease (AD). We previously reported that platelets contribute to Aß aggregation in cerebral vessels by secreting the factor clusterin upon binding of Aß40 to the fibrinogen receptor integrin αIIbß3 Here, we investigated the contribution of the collagen receptor GPVI (glycoprotein VI) in platelet-induced amyloid aggregation. Using platelets isolated from GPVI-wild type and GPVI-deficient human donors and mice, we found that Aß40 bound to GPVI, which induced the release of ATP and fibrinogen, resulting in platelet aggregation. Binding of Aß40 to integrin αIIbß3, fibrinogen, and GPVI collectively contributed to the formation of amyloid clusters at the platelet surface. Consequently, blockade of αIIbß3 or genetic loss of GPVI reduced amyloid fibril formation in cultured platelets and decreased the adhesion of Aß-activated platelets to injured carotid arteries in mice. Application of losartan to inhibit collagen binding to GPVI resulted in decreased Aß40-stimulated platelet activation, factor secretion, and platelet aggregation. Furthermore, the application of GPVI- or integrin-blocking antibodies reduced the formation of platelet-associated amyloid aggregates. Our findings indicate that Aß40 promotes platelet-mediated amyloid aggregation by binding to both GPVI and integrin αIIbß3 Blocking these pathways may therapeutically reduce amyloid plaque formation in cerebral vessels and the brain parenchyma of patients.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Plaquetas/metabolismo , Fragmentos de Peptídeos/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Agregação Patológica de Proteínas/metabolismo , Receptores de Colágeno/metabolismo , Adulto , Doença de Alzheimer/metabolismo , Animais , Plaquetas/citologia , Células Cultivadas , Fibrinogênio/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/genética , Ligação Proteica , Receptores de Colágeno/genética , Transdução de Sinais
11.
J Biol Chem ; 295(27): 9157-9170, 2020 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-32424040

RESUMO

C-type lectins that contain collagen-like domains are known as collectins. These proteins are present both in the circulation and in extravascular compartments and are central players of the innate immune system, contributing to first-line defenses against viral, bacterial, and fungal pathogens. The collectins mannose-binding lectin (MBL) and surfactant protein D (SP-D) are regulated by tissue fibroblasts at extravascular sites via an endocytic mechanism governed by urokinase plasminogen activator receptor-associated protein (uPARAP or Endo180), which is also a collagen receptor. Here, we investigated the molecular mechanisms that drive the uPARAP-mediated cellular uptake of MBL and SP-D. We found that the uptake depends on residues within a protruding loop in the fibronectin type-II (FNII) domain of uPARAP that are also critical for collagen uptake. Importantly, however, we also identified FNII domain residues having an exclusive role in collectin uptake. We noted that these residues are absent in the related collagen receptor, the mannose receptor (MR or CD206), which consistently does not interact with collectins. We also show that the second C-type lectin-like domain (CTLD2) is critical for the uptake of SP-D, but not MBL, indicating an additional level of complexity in the interactions between collectins and uPARAP. Finally, we demonstrate that the same molecular mechanisms enable uPARAP to engage MBL immobilized on the surface of pathogens, thereby expanding the potential biological implications of this interaction. Our study reveals molecular details of the receptor-mediated cellular regulation of collectins and offers critical clues for future investigations into collectin biology and pathology.


Assuntos
Colectinas/metabolismo , Endocitose/fisiologia , Receptores Mitogênicos/genética , Animais , Células CHO , Proteínas de Transporte/metabolismo , Colágeno/metabolismo , Cricetulus , Fibroblastos/metabolismo , Células HEK293 , Humanos , Lectinas Tipo C , Receptor de Manose , Lectina de Ligação a Manose/metabolismo , Lectinas de Ligação a Manose , Glicoproteínas de Membrana/metabolismo , Proteína D Associada a Surfactante Pulmonar/metabolismo , Receptores de Superfície Celular , Receptores de Colágeno/metabolismo , Receptores Mitogênicos/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo
12.
Dev Cell ; 53(4): 418-430.e4, 2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32428455

RESUMO

Capillary morphogenesis gene 2 (CMG2/ANTXR2) is a cell surface receptor for both collagen VI and anthrax toxin. Biallelic loss-of-function mutations in CMG2 lead to a severe condition, hyaline fibromatosis syndrome (HFS). We have here dissected a network of dynamic interactions between CMG2 and various actin interactors and regulators, describing a different behavior from other extracellular matrix receptors. CMG2 binds talin, and thereby the actin cytoskeleton, only in its ligand-free state. Extracellular ligand binding leads to src-dependent talin release and recruitment of the actin cytoskeleton regulator RhoA and its effectors. These sequential interactions of CMG2 are necessary for the control of oriented cell division during fish development. Finally, we demonstrate that effective switching between talin and RhoA binding is required for the intracellular degradation of collagen VI in human fibroblasts, which explains why HFS mutations in the cytoskeleton-binding domain lead to dysregulation of extracellular matrix homeostasis.


Assuntos
Colágeno Tipo VI/metabolismo , Endocitose , Receptores de Colágeno/metabolismo , Receptores de Peptídeos/metabolismo , Talina/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Citoesqueleto/metabolismo , Feminino , Humanos , /metabolismo , Ligantes , Masculino , Mutação , Receptores de Colágeno/genética , Receptores de Peptídeos/genética , Talina/genética , Peixe-Zebra , Proteína rhoA de Ligação ao GTP/genética
13.
Mol Cell Biochem ; 466(1-2): 1-15, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31912277

RESUMO

Progressive alteration of the extracellular matrix (ECM) is the characteristic of hypertensive nephropathy (HN). Both mesangial and endothelial cells have the ability to synthesize and degrade ECM components, including collagens through the activation of matrix metalloproteinases (MMPs) in stress conditions, such as in hypertension. On the other hand, hydrogen sulfide (H2S) has been shown to mitigate hypertensive renal matrix remodeling. Surprisingly, whether H2S ameliorates receptor-mediated (urokinase plasminogen activator receptor-associated protein, uPARAP/Endo180) collagen dysregulation in Ang-II hypertension is not clear. The purpose of this study was to determine whether Ang-II alters the expression of Endo180, tissue plasminogen activator (tPA), MMPs, and their tissue inhibitors (TIMPs) leading to the dysregulation of cellular collagen homeostasis and whether H2S mitigates the collagen turnover. Mouse mesangial cells (MCs) and glomerular endothelial cells (MGECs) were treated without or with Ang-II and H2S donor GYY (GYY4137) for 48 h. Cell lysates were analyzed by Western blot and RT-PCR, and cells were analyzed by immunocytochemistry. The results indicated that, while Ang-II differentially expressed MMP-13 and TIMP-1 in MCs and in MGECs, it predominantly decreased tPA, Endo 180, and increased plasminogen activator inhibitor-1 (PAI-1), MMP-14, and collagen IIIA and IV in both the cell types. Interestingly, H2S donor GYY treatment normalized the above changes in both the cell types. We conclude that Ang-II treatment causes ECM remodeling in MCs and MGECs through PAI-1/tPA/Endo180 and MMP/TIMP-dependent collagen remodeling, and H2S treatment mitigates remodeling, in part, by modulating these pathways.


Assuntos
Células Endoteliais/metabolismo , Mesângio Glomerular/metabolismo , Hipertensão Renal/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Receptores de Colágeno/metabolismo , Estresse Fisiológico , Animais , Células Endoteliais/patologia , Mesângio Glomerular/patologia , Hipertensão Renal/patologia , Camundongos
14.
J Biol Chem ; 294(51): 19723-19739, 2019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31699892

RESUMO

Excessive collagen deposition by myofibroblasts during adverse cardiac remodeling leads to myocardial fibrosis that can compromise cardiac function. Unraveling the mechanisms underlying collagen gene expression in cardiac myofibroblasts is therefore an important clinical goal. The collagen receptors, discoidin domain receptor 2 (DDR2), a collagen-specific receptor tyrosine kinase, and integrin-ß1, are reported to mediate tissue fibrosis. Here, we probed the role of DDR2-integrin-ß1 cross-talk in the regulation of collagen α1(I) gene expression in angiotensin II (Ang II)-stimulated cardiac fibroblasts. Results from gene silencing/overexpression approaches, electrophoretic mobility shift assays, and ChIP revealed that DDR2 acts via extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase (ERK1/2 MAPK)-dependent transforming growth factor-ß1 (TGF-ß1) signaling to activate activator protein-1 (AP-1) that in turn transcriptionally enhances the expression of collagen-binding integrin-ß1 in Ang II-stimulated cardiac fibroblasts. The DDR2-integrin-ß1 link was also evident in spontaneously hypertensive rats and DDR2-knockout mice. Further, DDR2 acted via integrin-ß1 to regulate α-smooth muscle actin (α-SMA) and collagen type I expression in Ang II-exposed cardiac fibroblasts. Downstream of the DDR2-integrin-ß1 axis, α-SMA was found to regulate collagen α1(I) gene expression via the Ca2+ channel, transient receptor potential cation channel subfamily C member 6 (TRPC6), and the profibrotic transcription factor, Yes-associated protein (YAP). This finding indicated that fibroblast-to-myofibroblast conversion is mechanistically coupled to collagen expression. The observation that collagen receptor cross-talk underlies α-SMA-dependent collagen type I expression in cardiac fibroblasts expands our understanding of the complex mechanisms involved in collagen gene expression in the heart and may be relevant to cardiac fibrogenesis.


Assuntos
Actinas/metabolismo , Angiotensina II/farmacologia , Fibroblastos/metabolismo , Miocárdio/citologia , Receptores de Colágeno/metabolismo , Animais , Colágeno/metabolismo , Meios de Cultivo Condicionados , Fibroblastos/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Integrina beta1/metabolismo , Integrinas/metabolismo , Masculino , Camundongos , Camundongos Knockout , Músculo Liso/metabolismo , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Canais de Cátion TRPC/metabolismo , Canal de Cátion TRPC6/metabolismo
15.
Blood Rev ; 38: 100592, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31351674

RESUMO

Much interest surrounds the receptors α2ß1 and glycoprotein VI (GPVI) whose synchronized action mediates the attachment and activation of platelets on collagen, essential for preventing blood loss but also the most thrombogenic component of the vessel wall. Subject to density variations on platelets through natural polymorphisms, the absence of α2ß1 or GPVI uniquely leads to a substantial block of hemostasis without causing major bleeding. Specific to the megakaryocyte lineage, GPVI and its signaling pathways are most promising targets for anti-thrombotic therapy. This review looks at the clinical consequences of the loss of collagen receptor function with emphasis on both the inherited and acquired loss of GPVI with brief mention of mouse models when necessary. A detailed survey of rare case reports of patients with inherited disease-causing variants of the GP6 gene is followed by an assessment of the causes and clinical consequences of acquired GPVI deficiency, a more frequent finding most often due to antibody-induced platelet GPVI shedding. Release of soluble GPVI is brought about by platelet metalloproteinases; a process induced by ligand or antibody binding to GPVI or even high shear forces. Also included is an assessment of the clinical importance of GPVI-mediated platelet interactions with fibrin and of the promise shown by the pharmacological inhibition of GPVI in a cardiovascular context. The role for GPVI in platelet function in inflammation and in the evolution and treatment of major illnesses such as rheumatoid arthritis, cancer and sepsis is also discussed.


Assuntos
Colágeno/metabolismo , Ativação Plaquetária , Glicoproteínas da Membrana de Plaquetas/metabolismo , Animais , Plaquetas/metabolismo , Humanos , Integrina alfa2beta1/metabolismo , Receptores de Colágeno/metabolismo , Transdução de Sinais
16.
Biochim Biophys Acta Mol Cell Res ; 1866(11): 118496, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31229648

RESUMO

Assembly of cell-surface receptors into specific oligomeric states and/or clusters before and after ligand binding is an important feature governing their biological function. Receptor oligomerization can be mediated by specific domains of the receptor, ligand binding, configurational changes or other interacting molecules. In this review we summarize our understanding of the oligomeric state of discoidin domain receptors (DDR1 and DDR2), which belong to the receptor tyrosine kinase family (RTK). DDRs form an interesting system from an oligomerization perspective as their ligand collagen(s) can also undergo supramolecular assembly to form fibrils. Even though DDR1 and DDR2 differ in the domains responsible to form ligand-free dimers they share similarities in binding to soluble, monomeric collagen. However, only DDR1b forms globular clusters in response to monomeric collagen and not DDR2. Interestingly, both DDR1 and DDR2 are assembled into linear clusters by the collagen fibril. Formation of these clusters is important for receptor phosphorylation and is mediated in part by other membrane components. We summarize how the oligomeric status of DDRs shares similarities with other members of the RTK family and with collagen receptors. Unraveling the multiple macro-molecular configurations adopted by this receptor-ligand pair can provide novel insights into the intricacies of cell-matrix interactions.


Assuntos
Receptores com Domínio Discoidina/química , Receptores com Domínio Discoidina/metabolismo , Ligação Proteica , Sítios de Ligação , Colágeno/química , Domínio Discoidina , Receptor com Domínio Discoidina 1/química , Receptor com Domínio Discoidina 1/metabolismo , Receptor com Domínio Discoidina 2/química , Receptor com Domínio Discoidina 2/metabolismo , Colágenos Fibrilares , Humanos , Ligantes , Fosforilação , Receptores Proteína Tirosina Quinases , Receptores de Colágeno/química , Receptores de Colágeno/metabolismo
17.
Biochim Biophys Acta Mol Cell Res ; 1866(11): 118478, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31004686

RESUMO

The structure of the collagen fibril surface directly effects and possibly assists the management of collagen receptor interactions. An important class of collagen receptors, the receptor tyrosine kinases of the Discoidin Domain Receptor family (DDR1 and DDR2), are differentially activated by specific collagen types and play important roles in cell adhesion, migration, proliferation, and matrix remodeling. This review discusses their structure and function as it pertains directly to the fibrillar collagen structure with which they interact far more readily than they do with isolated molecular collagen. This prospective provides further insight into the mechanisms of activation and rational cellular control of this important class of receptors while also providing a comparison of DDR-collagen interactions with other receptors such as integrin and GPVI. When improperly regulated, DDR activation can lead to abnormal cellular proliferation activities such as in cancer. Hence how and when the DDRs associate with the major basis of mammalian tissue infrastructure, fibrillar collagen, should be of keen interest.


Assuntos
Colágeno/metabolismo , Receptores com Domínio Discoidina/metabolismo , Ligação Proteica , Receptores de Colágeno/metabolismo , Animais , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Colágeno/química , Receptor com Domínio Discoidina 1 , Receptor com Domínio Discoidina 2 , Receptores com Domínio Discoidina/química , Colágenos Fibrilares/química , Colágenos Fibrilares/metabolismo , Humanos , Ligantes , Modelos Moleculares , Estrutura Molecular , Neoplasias/metabolismo , Domínios e Motivos de Interação entre Proteínas , Receptores Proteína Tirosina Quinases , Receptores de Colágeno/química , Transdução de Sinais
18.
Cells ; 8(4)2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30939867

RESUMO

A hallmark of ageing is the redistribution of body fat. Particularly, subcutaneous fat decreases paralleled by a decrease of skin collagen I are typical for age-related skin atrophy. In this paper, we hypothesize that collagen I may be a relevant molecule stimulating the differentiation of adipose-derived stem cells (ASCs) into adipocytes augmenting subcutaneous fat. In this context lipogenesis, adiponectin, and collagen I receptor expression were determined. Freshly isolated ASCs were characterized by stemness-associated surface markers by FACS analysis and then transdifferentiated into adipocytes by specific medium supplements. Lipogenesis was evaluated using Nile Red staining and documented by fluorescence microscopy or quantitatively measured by using a multiwell spectrofluorometer. Expression of adiponectin was measured by real-time RT-PCR and in cell-free supernatants by ELISA, and expression of collagen I receptors was observed by western blot analysis. It was found that supports coated with collagen I promote cell adhesion and lipogenesis of ASCs. Interestingly, a reverse correlation to adiponectin expression was observed. Moreover, we found upregulation of the collagen receptor, discoidin domain-containing receptor 2; receptors of the integrin family were absent or downregulated. These findings indicate that collagen I is able to modulate lipogenesis and adiponectin expression and therefore may contribute to metabolic dysfunctions associated with ageing.


Assuntos
Adipócitos/citologia , Tecido Adiposo/citologia , Colágeno Tipo I/farmacologia , Células-Tronco/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adiponectina/genética , Adiponectina/metabolismo , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Integrina alfa2beta1/metabolismo , Lipogênese/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Colágeno/metabolismo , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
19.
BMC Cancer ; 19(1): 234, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-30876468

RESUMO

BACKGROUND: Cancer progression is influenced by a pro-tumorigenic microenvironment. The aberrant tumor stroma with increased collagen deposition, contractile fibroblasts and dysfunctional vessels has a major impact on the interstitial fluid pressure (PIF) in most solid tumors. An increased tumor PIF is a barrier to the transport of interstitial fluid into and within the tumor. Therefore, understanding the mechanisms that regulate pressure homeostasis can lead to new insight into breast tumor progression, invasion and response to therapy. The collagen binding integrin α11ß1 is upregulated during myofibroblast differentiation and expressed on fibroblasts in the tumor stroma. As a collagen organizer and a probable link between contractile fibroblasts and the complex collagen network in tumors, integrin α11ß1 could be a potential regulator of tumor PIF. METHODS: We investigated the effect of stromal integrin α11-deficiency on pressure homeostasis, collagen organization and tumor growth using orthotopic and ectopic triple-negative breast cancer xenografts (MDA-MB-231 and MDA-MB-468) in wild type and integrin α11-deficient mice. PIF was measured by the wick-in-needle technique, collagen by Picrosirius Red staining and electron microscopy, and uptake of radioactively labeled 5FU by microdialysis. Further, PIF in heterospheroids composed of MDA-MB-231 cells and wild type or integrin α11-deficient fibroblasts was measured by micropuncture. RESULTS: Stromal integrin α11-deficiency decreased PIF in both the orthotopic breast cancer models. A concomitant perturbed collagen structure was seen, with fewer aligned and thinner fibrils. Integrin α11-deficiency also impeded MDA-MB-231 breast tumor growth, but no effect was observed on drug uptake. No effects were seen in the ectopic model. By investigating the isolated effect of integrin α11-positive fibroblasts on MDA-MB-231 cells in vitro, we provide evidence that PIF regulation was mediated by integrin α11-positive fibroblasts. CONCLUSION: We hereby show the importance of integrin α11ß1 in pressure homeostasis in triple-negative breast tumors, indicating a new role for integrin α11ß1 in the tumor microenvironment. Our data suggest that integrin α11ß1 has a pro-tumorigenic effect on triple-negative breast cancer growth in vivo. The significance of the local microenvironment is shown by the different effects of integrin α11ß1 in the orthotopic and ectopic models, underlining the importance of choosing an appropriate preclinical model.


Assuntos
Colágeno/química , Líquido Extracelular/metabolismo , Cadeias alfa de Integrinas/genética , Integrinas/metabolismo , Receptores de Colágeno/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Animais , Linhagem Celular Tumoral , Citoproteção , Feminino , Técnicas de Inativação de Genes , Humanos , Camundongos , Transplante de Neoplasias , Células Estromais , Neoplasias de Mama Triplo Negativas/química , Neoplasias de Mama Triplo Negativas/genética , Microambiente Tumoral
20.
Tissue Eng Part A ; 25(5-6): 390-398, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30569829

RESUMO

IMPACT STATEMENT: Complex and functional artificial tissues consisting of multiple types of cells are generally required. However, few reliable methods to control the cellular distribution have been developed. Our present study has revealed how the core-shell type distribution is formed in NIH3T3/MIN6 spheroids. We demonstrated that focal adhesion kinase signal was the key for the cell localization in the spheroids. Moreover, we succeeded in regulating their distribution based on the mechanism revealed in the present study. These novel findings will provide a new approach for constructing artificial tissues with proper cell arrangement, which would be suitable for tissue engineering.


Assuntos
Movimento Celular , Esferoides Celulares/citologia , Animais , Movimento Celular/efeitos dos fármacos , Colágeno Tipo I/farmacologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Células Hep G2 , Humanos , Integrinas/metabolismo , Camundongos , Receptores de Colágeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...