Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 710
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 3080, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35197532

RESUMO

Fibrosis is a leading cause of morbidity and mortality worldwide. Although fibrosis may involve different organ systems, transforming growth factor-ß (TGFß) has been established as a master regulator of fibrosis across organs. Pirfenidone and Nintedanib are the only currently-approved drugs to treat fibrosis, specifically idiopathic pulmonary fibrosis, but their mechanisms of action remain poorly understood. To identify novel drug targets and uncover potential mechanisms by which these drugs attenuate fibrosis, we performed an integrative 'omics analysis of transcriptomic and proteomic responses to TGFß1-stimulated lung fibroblasts. Significant findings were annotated as associated with pirfenidone and nintedanib treatment in silico via Coremine. Integrative 'omics identified a co-expressed transcriptomic and proteomic module significantly correlated with TGFß1 treatment that was enriched (FDR-p = 0.04) with genes associated with pirfenidone and nintedanib treatment. While a subset of genes in this module have been implicated in fibrogenesis, several novel TGFß1 signaling targets were identified. Specifically, four genes (BASP1, HSD17B6, CDH11, and TNS1) have been associated with pirfenidone, while five genes (CLINT1, CADM1, MTDH, SYDE1, and MCTS1) have been associated with nintedanib, and MYDGF has been implicated with treatment using both drugs. Using the Clue Drug Repurposing Hub, succinic acid was highlighted as a metabolite regulated by the protein encoded by HSD17B6. This study provides new insights into the anti-fibrotic actions of pirfenidone and nintedanib and identifies novel targets for future mechanistic studies.


Assuntos
Antifibróticos/farmacologia , Biologia Computacional/métodos , Proteínas da Matriz Extracelular/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/patologia , Indóis/farmacologia , Piridonas/farmacologia , Fator de Crescimento Transformador beta/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Antifibróticos/uso terapêutico , Caderinas/genética , Caderinas/metabolismo , Molécula 1 de Adesão Celular/genética , Molécula 1 de Adesão Celular/metabolismo , Feminino , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Indóis/uso terapêutico , Interleucinas/genética , Interleucinas/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Piridonas/uso terapêutico , Racemases e Epimerases/genética , Racemases e Epimerases/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Tensinas/genética , Tensinas/metabolismo
2.
Gene ; 815: 146137, 2022 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-35007686

RESUMO

The extracellular matrix (ECM) is composed of a mesh of proteins, proteoglycans, growth factors, and other secretory components. It constitutes the tumor microenvironment along with the endothelial cells, cancer-associated fibroblasts, adipocytes, and immune cells. The proteins of ECM can be functionally classified as adhesive proteins and matricellular proteins (MCP). In the tumor milieu, the ECM plays a major role in tumorigenesis and therapeutic resistance. The current review encompasses thrombospondins, osteonectin, osteopontin, tenascin C, periostin, the CCN family, laminin, biglycan, decorin, mimecan, and galectins. The matrix metalloproteinases (MMPs) are also discussed as they are an integral part of the ECM with versatile functions in the tumor stroma. In this review, the role of these proteins in tumor initiation, growth, invasion and metastasis have been highlighted, with emphasis on their contribution to tumor therapeutic resistance. Further, their potential as biomarkers and therapeutic targets based on existing evidence are discussed. Owing to the recent advancements in protein targeting, the possibility of agents to modulate MCPs in cancer as therapeutic options are discussed.


Assuntos
Biomarcadores Tumorais , Proteínas da Matriz Extracelular/fisiologia , Neoplasias/etiologia , Neoplasias/terapia , Moléculas de Adesão Celular/fisiologia , Proteínas da Matriz Extracelular/análise , Humanos , Metaloproteinases da Matriz/fisiologia , Osteonectina/análise , Osteonectina/fisiologia , Osteopontina/fisiologia , Tenascina/fisiologia , Trombospondina 1/fisiologia , Resultado do Tratamento
3.
Int J Mol Sci ; 22(12)2021 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-34205668

RESUMO

Matricellular proteins (MCPs) are defined as extracellular matrix (ECM) associated proteins that are important regulators and integrators of microenvironmental signals, contributing to the dynamic nature of ECM signalling. There is a growing understanding of the role of matricellular proteins in cellular processes governing tissue development as well as in disease pathogenesis. In this review, the expression and functions of different MP family members (periostin, CCNs, TSPs, SIBLINGs and others) are presented, specifically in relation to craniofacial development and the maintenance of orofacial tissues, including bone, gingiva, oral mucosa, palate and the dental pulp. As will be discussed, each MP family member has been shown to have non-redundant roles in development, tissue homeostasis, wound healing, pathology and tumorigenesis of orofacial and dental tissues.


Assuntos
Moléculas de Adesão Celular/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Boca/crescimento & desenvolvimento , Osteonectina/fisiologia , Trombospondinas/fisiologia , Animais , Proteínas de Sinalização Intercelular CCN/fisiologia , Neoplasias de Cabeça e Pescoço/etiologia , Humanos , Boca/embriologia , Tenascina/fisiologia , Cicatrização
4.
Med Sci Monit ; 27: e931238, 2021 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-34210950

RESUMO

BACKGROUND Oral squamous cell carcinoma (OSCC) is a common tumor of the head and neck. Its treatment usually requires multiple modalities. Currently, there are no molecular biomarkers to guide these treatment strategies. Studies have shown that microfibril-associated protein 4 (MFAP4) is potentially useful for non-invasive assessment of various diseases; however, its biological function in tumors is still unknown. In this study, we propose that MFAP4 is a new prognostic target for OSCC. MATERIAL AND METHODS First, we collected OSCC data (GSE25099 and GSE30784 datasets) from the Gene Expression Omnibus (GEO) database and compared the differential expression of MFAP4 gene between the patients (tumor) and normal (control) groups. The comparison was done with University of California Santa Cruz Xena (https://xenabrowser.net/Datapages/), and we calculated the difference in MFAP4 gene expression between normal and tumor tissues in a pan-cancer analysis. Then, we compared the 2 groups with high and low expression of MFAP4 gene in terms of tumor mutation burden (TMB), miRNA regulation, and immune cell infiltration. RESULTS We found that the expression of MFAP4 gene was significantly decreased in tumors. Our research also showed that high expression of MFAP4 was related to better prognosis of patients and may be related to tumor gene mutation, miRNA regulation, and infiltration of different immune cells. CONCLUSIONS Our work provides evidence that expression of MFAP4 can be used as a prognostic biomarker for risk stratification of OSCC patients and elaborates on its relation with the regulation of TMB, miRNAs, and immune cell infiltration.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Glicoproteínas/fisiologia , Neoplasias Bucais/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Biomarcadores Tumorais/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos
5.
Mol Cell Neurosci ; 112: 103614, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33845123

RESUMO

Homozygous Dab1 yotari mutant mice, Dab1yot (yot/yot) mice, have an autosomal recessive mutation of Dab1 and show reeler-like phenotype including histological abnormality of the cerebellum, hippocampus, and cerebral cortex. We here show abnormal hippocampal development of yot/yot mice where granule cells and pyramidal cells fail to form orderly rows but are dispersed diffusely in vague multiplicative layers. Possibly due to the positioning failure of granule cells and pyramidal cells and insufficient synaptogenesis, axons of the granule cells did not extend purposefully to connect with neighboring regions in yot/yot mice. We found that both hippocampal granule cells and pyramidal cells of yot/yot mice expressed proteins reactive with the anti-Dab1 antibody. We found that Y198- phosphorylated Dab1 of yot/yot mice was greatly decreased. Accordingly the downstream molecule, Akt was hardly phosphorylated. Especially, synapse formation was defective and the distribution of neurons was scattered in hippocampus of yot/yot mice. Some of neural cell adhesion molecules and hippocampus associated transcription factors of the neurons were expressed aberrantly, suggesting that the Reelin-Dab1 signaling pathway seemed to be importantly involved in not only neural migration as having been shown previously but also neural maturation and/or synaptogenesis of the mice. It is interesting to clarify whether the defective neural maturation is a direct consequence of the dysfunctional Dab1, or alternatively secondarily due to the Reelin-Dab1 intracellular signaling pathways.


Assuntos
Moléculas de Adesão Celular Neuronais/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Hipocampo/anormalidades , Camundongos Mutantes/anormalidades , Proteínas do Tecido Nervoso/fisiologia , Serina Endopeptidases/fisiologia , Transdução de Sinais/fisiologia , Animais , Moléculas de Adesão Celular Neuronais/deficiência , Movimento Celular , Ativação Enzimática , Proteínas da Matriz Extracelular/deficiência , Genes Recessivos , Hipocampo/embriologia , Hipocampo/metabolismo , Hipocampo/patologia , Homozigoto , Camundongos , Camundongos Mutantes/genética , Camundongos Mutantes/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Moléculas de Adesão de Célula Nervosa/biossíntese , Moléculas de Adesão de Célula Nervosa/genética , Fenótipo , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína Reelina , Serina Endopeptidases/deficiência , Sinapses/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
6.
Int J Mol Sci ; 22(5)2021 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-33670905

RESUMO

Rheumatoid arthritis (RA) is a chronic autoimmune disease causing inflammation of joints, cartilage destruction and bone erosion. Biomarkers and new drug targets are actively sought and progressed to improve available options for patient treatment. The Collagen Triple Helix Repeat Containing 1 protein (CTHRC1) may have an important role as a biomarker for rheumatoid arthritis, as CTHRC1 protein concentration is significantly elevated in the peripheral blood of rheumatoid arthritis patients compared to osteoarthritis (OA) patients and healthy individuals. CTHRC1 is a secreted glycoprotein that promotes cell migration and has been implicated in arterial tissue-repair processes. Furthermore, high CTHRC1 expression is observed in many types of cancer and is associated with cancer metastasis to the bone and poor patient prognosis. However, the function of CTHRC1 in RA is still largely undefined. The aim of this review is to summarize recent findings on the role of CTHRC1 as a potential biomarker and pathogenic driver of RA progression. We will discuss emerging evidence linking CTHRC1 to the pathogenic behavior of fibroblast-like synoviocytes and to cartilage and bone erosion through modulation of the balance between bone resorption and repair.


Assuntos
Artrite Reumatoide/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Animais , Artrite Reumatoide/fisiopatologia , Biomarcadores , Proteínas da Matriz Extracelular/fisiologia , Humanos , Sinoviócitos/metabolismo , Sinoviócitos/fisiologia , Via de Sinalização Wnt
7.
Dev Dyn ; 250(2): 274-294, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33012048

RESUMO

BACKGROUND: The extracellular matrix (ECM) is a fundamental component of multicellular organisms that orchestrates developmental processes and controls cell and tissue organization. We previously identified the novel ECM protein SNED1 as a promoter of breast cancer metastasis and showed that its level of expression negatively correlated with breast cancer patient survival. Here, we sought to identify the roles of SNED1 during murine development. RESULTS: We generated two novel Sned1 knockout mouse strains and showed that Sned1 is essential since homozygous ablation of the gene led to early neonatal lethality. Phenotypic analysis of the surviving knockout mice revealed a role for SNED1 in the development of craniofacial and skeletal structures since Sned1 knockout resulted in growth defects, nasal cavity occlusion, and craniofacial malformations. Sned1 is widely expressed in embryos, notably by cell populations undergoing epithelial-to-mesenchymal transition, such as the neural crest cells. We further show that mice with a neural-crest-cell-specific deletion of Sned1 survive, but display facial anomalies partly phenocopying the global knockout mice. CONCLUSIONS: Our results demonstrate requisite roles for SNED1 during development and neonatal survival. Importantly, the deletion of 2q37.3 in humans, a region that includes the SNED1 locus, has been associated with facial dysmorphism and short stature.


Assuntos
Proteínas da Matriz Extracelular/fisiologia , Sequência de Aminoácidos , Animais , Sequência Conservada , Anormalidades Craniofaciais/genética , Genes Letais , Transtornos do Crescimento/genética , Mandíbula/anormalidades , Camundongos , Camundongos Knockout , Cavidade Nasal/anormalidades
8.
J Gastroenterol Hepatol ; 36(1): 196-203, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32537806

RESUMO

BACKGROUND AND AIM: Tubulointerstitial nephritis antigen-like 1 (TINAGL1), as a novel matricellular protein, has been demonstrated to participate in cancer progression, whereas the potential function of TINAGL1 in gastric cancer (GC) remains unknown. METHODS: The expression pattern of TINAGL1 in GC was examined by immunohistochemistry, ELISA, real-time polymerase chain reaction, and Western blot. Correlation between TINAGL1 and matrix metalloproteinases (MMPs) was analyzed by the GEPIA website and Kaplan-Meier plots database. The lentivirus-based TINAGL1 knockdown, CCK-8, and transwell assays were used to test the function of TINAGL1 in vitro. The role of TINAGL1 was confirmed by subcutaneous xenograft, abdominal dissemination, and lung metastasis model. Microarray experiments, ELISA, real-time polymerase chain reaction, and Western blot were used to identify molecular mechanism. RESULTS: TINAGL1 was increased in GC tumor tissues and associated with poor patient survival. Moreover, TINAGL1 significantly promoted GC cell proliferation and migration in vitro as well as facilitated GC tumor growth and metastasis in vivo. TINAGL1 expression in GC cells was accompanied with increasing MMPs including MMP2, MMP9, MMP11, MMP14, and MMP16. GEPIA database revealed that these MMPs were correlated with TINAGL1 in GC tumors and that the most highly expressed MMP was MMP2. Mechanically, TINAGL1 regulated MMP2 through the JNK signaling pathway activation. CONCLUSIONS: Our data highlight that TINAGL1 promotes GC growth and metastasis and regulates MMP2 expression, indicating that TINAGL1 may serve as a therapeutic target for GC.


Assuntos
Proliferação de Células/genética , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Expressão Gênica/genética , Lipocalinas/genética , Lipocalinas/metabolismo , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Metástase Neoplásica/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Regulação para Cima/genética , Regulação para Cima/fisiologia , Animais , Linhagem Celular , Movimento Celular/genética , Modelos Animais de Doenças , Progressão da Doença , Proteínas da Matriz Extracelular/fisiologia , Feminino , Humanos , Lipocalinas/fisiologia , Camundongos Nus , Terapia de Alvo Molecular , Neoplasias Gástricas/terapia
9.
Biosci Rep ; 41(1)2021 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33306121

RESUMO

BACKGROUND: The family with sequence similarity 20-member C (Fam20C) kinase plays important roles in physiopathological process and is responsible for majority of the secreted phosphoproteome, including substrates associated with tumor cell migration. However, it remains unclear whether Fam20C plays a role in cancers. Here, we aimed to analyze the expression and prognostic value of Fam20C in pan-cancer and to gain insights into the association between Fam20C and immune infiltration. METHODS: We analyzed Fam20C expression patterns and the associations between Fam20C expression levels and prognosis in pan-cancer via the ONCOMINE, TIMER (Tumor Immune Estimation Resource), PrognoScan, GEPIA (Gene Expression Profiling Interactive Analysis), and Kaplan-Meier Plotter databases. After that, GEPIA and TIMER databases were applied to investigate the relations between Fam20C expression and immune infiltration across different cancer types, especially BLCA (bladder urothelial carcinoma), LGG (brain lower grade glioma), and STAD (stomach adenocarcinoma). RESULTS: Compared with adjacent normal tissues, Fam20C was widely expressed across many cancers. In general, Fam20C showed a detrimental role in pan-cancer, it was positively associated with poor survival of BLCA, LGG, and STAD patients. Specifically, based on TCGA (The Cancer Genome Atlas) database, a high expression level of Fam20C was associated with worse prognostic value in stages T2-T4 and stages N0-N2 in the cohort of STAD patients. Moreover, Fam20C expression had positive associations with immune infiltration, including CD4+ T cells, macrophages, neutrophils, and dendritic cells, and other diverse immune cells in BLCA, LGG, and STAD. CONCLUSION: Fam20C may serve as a promising prognostic biomarker in pan-cancer and has positive associations with immune infiltrates.


Assuntos
Caseína Quinase I/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Neoplasias/imunologia , Caseína Quinase I/genética , Proteínas da Matriz Extracelular/genética , Humanos , Estimativa de Kaplan-Meier , Metástase Linfática , Neoplasias/genética , Neoplasias/patologia , Oncogenes , Prognóstico , RNA Mensageiro/genética
10.
Sci Rep ; 10(1): 18583, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-33122788

RESUMO

The ability to ablate a gene in a given tissue by generating a conditional knockout (cKO) is crucial for determining its function in the targeted tissue. Such tissue-specific ablation is even more critical when the gene's conventional knockout (KO) is lethal, which precludes studying the consequences of its deletion in other tissues. Therefore, here we describe a successful strategy that generated a Matrix Gla floxed mouse (Mgp.floxed) by the CRISPR/Cas9 system, that subsequently allowed the generation of cKOs by local viral delivery of the Cre-recombinase enzyme. MGP is a well-established inhibitor of calcification gene, highly expressed in arteries' smooth muscle cells and chondrocytes. MGP is also one of the most abundant genes in the trabecular meshwork, the eye tissue responsible for maintenance of intraocular pressure (IOP) and development of Glaucoma. Our strategy entailed one-step injection of two gRNAs, Cas9 protein and a long-single-stranded-circular DNA donor vector (lsscDNA, 6.7 kb) containing two loxP sites in cis and 900-700 bp 5'/3' homology arms. Ocular intracameral injection of Mgp.floxed mice with a Cre-adenovirus, led to an Mgp.TMcKO mouse which developed elevated IOP. Our study discovered a new role for the Mgp gene as a keeper of physiological IOP in the eye.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Olho/fisiopatologia , Pressão Intraocular , Malha Trabecular/fisiopatologia , Animais , Sequência de Bases , Feminino , Glaucoma/fisiopatologia , Integrases/metabolismo , Camundongos , Camundongos Knockout , RNA Guia de Cinetoplastídeos/administração & dosagem , Proteína de Matriz Gla
11.
Biomolecules ; 10(9)2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32911658

RESUMO

Fibulin-3 (also known as EGF-containing fibulin extracellular matrix protein 1 (EFEMP1)) is a secreted extracellular matrix glycoprotein, encoded by the EFEMP1 gene that belongs to the eight-membered fibulin protein family. It has emerged as a functionally unique member of this family, with a diverse array of pathophysiological associations predominantly centered on its role as a modulator of extracellular matrix (ECM) biology. Fibulin-3 is widely expressed in the human body, especially in elastic-fibre-rich tissues and ocular structures, and interacts with enzymatic ECM regulators, including tissue inhibitor of metalloproteinase-3 (TIMP-3). A point mutation in EFEMP1 causes an inherited early-onset form of macular degeneration called Malattia Leventinese/Doyne honeycomb retinal dystrophy (ML/DHRD). EFEMP1 genetic variants have also been associated in genome-wide association studies with numerous complex inherited phenotypes, both physiological (namely, developmental anthropometric traits) and pathological (many of which involve abnormalities of connective tissue function). Furthermore, EFEMP1 expression changes are implicated in the progression of numerous types of cancer, an area in which fibulin-3 has putative significance as a therapeutic target. Here we discuss the potential mechanistic roles of fibulin-3 in these pathologies and highlight how it may contribute to the development, structural integrity, and emergent functionality of the ECM and connective tissues across a range of anatomical locations. Its myriad of aetiological roles positions fibulin-3 as a molecule of interest across numerous research fields and may inform our future understanding and therapeutic approach to many human diseases in clinical settings.


Assuntos
Proteínas da Matriz Extracelular/fisiologia , Animais , Modelos Animais de Doenças , Matriz Extracelular/enzimologia , Proteínas da Matriz Extracelular/química , Estudo de Associação Genômica Ampla , Humanos , Drusas do Disco Óptico/congênito , Drusas do Disco Óptico/genética , Drusas do Disco Óptico/fisiopatologia
12.
Med Sci Monit ; 26: e925298, 2020 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-32764530

RESUMO

BACKGROUND Emerging evidence suggests the involvement of Reelin in chemoresistance in various cancers. However, its function in cisplatin (DDP) sensitivity of non-small cell lung cancer (NSCLC) needs to be investigated. MATERIAL AND METHODS Reelin expression in cisplatin-sensitive A549 cells and cisplatin-resistant NSCLC (A549/DDP) cells was analyzed by western blot analysis. qRT-PCR, western blotting, immunofluorescence, CCK-8 assays, Annexin V/propidium iodide apoptosis assay, and Transwell migration assays were carried out to determine the function of Reelin on DDP resistance. RESULTS Reelin was markedly increased in A549/DDP cells relative to A549 cells. Knockdown of Reelin enhanced DDP chemosensitivity of A549/DDP cells, whereas overexpression of Reelin enhanced DDP resistance of A549, H1299, and H460 cells. Reelin induced DDP resistance in NSCLC cells via facilitating epithelial-mesenchymal transition (EMT). Furthermore, Reelin modulated p38/GSK3ß signal transduction and promoted Snail (EMT-associated transcription factor) expression. Suppression of p38/Snail reversed Reelin-induced EMT and resistance of NSCLC cells to DDP. CONCLUSIONS These data indicated that Reelin induces DDP resistance of NSCLC by regulation of the p38/GSK3ß/Snail/EMT signaling pathway and provide evidence that Reelin suppression can be an effective strategy to suppress DDP resistance in NSCLC.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Moléculas de Adesão Celular Neuronais/fisiologia , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Proteínas do Tecido Nervoso/fisiologia , Serina Endopeptidases/fisiologia , Transdução de Sinais/fisiologia , Fatores de Transcrição da Família Snail/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Proteína Reelina
13.
Mol Cell Endocrinol ; 517: 110931, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32712387

RESUMO

The proteolytic fragment ASARM (acidic serine- and aspartate-rich motif) of MEPE (matrix extracellular phosphoglycoprotein) (MEPE-ASARM) may act as an endogenous anti-mineralization factor involved in X-linked hypophosphatemic rickets/osteomalacia (XLH). We synthesized MEPE-ASARM peptides and relevant peptide fragments with or without phosphorylated Ser residues (pSer) to determine the active site(s) of MEPE-ASARM in a rat calvaria cell culture model. None of the synthetic peptides elicited changes in cell death, proliferation or differentiation, but the peptide (pASARM) with three pSer residues inhibited mineralization without causing changes in gene expression of osteoblast markers tested. The anti-mineralization effect was maintained in peptides in which any one of three pSer residues was deleted. Polyclonal antibodies recognizing pASARM but not ASARM abolished the pASARM effect. Deletion of six N-terminal residues but leaving the recognition sites for PHEX (phosphate regulating endopeptidase homolog, X-linked), a membrane endopeptidase responsible for XLH, intact and two C-terminal amino acid residues did not alter the anti-mineralization activity of pASARM. Our results strengthen understanding of the active sites of MEPE-pASARM and allowed us to identify a shorter more stable sequence with fewer pSer residues still exhibiting hypomineralization activity, reducing peptide synthesis cost and increasing reliability for exploring biological and potential therapeutic effects.


Assuntos
Matriz Óssea/metabolismo , Calcificação Fisiológica/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Glicoproteínas/fisiologia , Fragmentos de Peptídeos/fisiologia , Fosfoproteínas/fisiologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/imunologia , Especificidade de Anticorpos , Domínio Catalítico , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/imunologia , Glicoproteínas/química , Glicoproteínas/imunologia , Humanos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Endopeptidase Neutra Reguladora de Fosfato PHEX , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/farmacologia , Fosfoproteínas/química , Fosfoproteínas/imunologia , Fosforilação , Fosfosserina/análise , Processamento de Proteína Pós-Traducional , Coelhos , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Crânio/citologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
14.
Calcif Tissue Int ; 107(2): 180-190, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32462291

RESUMO

Microgravity causes both muscle and bone loss. Although we previously revealed that gravity change influences muscle and bone through the vestibular system in mice, its detailed mechanism has not been elucidated. In this study, we investigated the roles of olfactomedin 1 (OLFM1), whose expression was upregulated during hypergravity in the soleus muscle, in mouse bone cells. Vestibular lesion significantly blunted OLFM1 expression in the soleus muscle and serum OLFM1 levels enhanced by hypergravity in mice. Moreover, a phosphatidylinositol 3-kinase inhibitor antagonized shear stress-enhanced OLFM1 expression in C2C12 myotubes. As for the effects of OLFM1 on bone cells, OLFM1 inhibited osteoclast formation from mouse bone marrow cells and mouse preosteoclastic RAW264.7 cells. Moreover, OLFM1 suppressed RANKL expression and nuclear factor-κB signaling in mouse osteoblasts. Serum OLFM1 levels were positively related to OLFM1 mRNA levels in the soleus muscle and trabecular bone mineral density of mice. In conclusion, we first showed that OLFM1 suppresses osteoclast formation and RANKL expression in mouse cells.


Assuntos
Osso e Ossos/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Glicoproteínas/fisiologia , Hipergravidade , Músculo Esquelético/fisiologia , Animais , Diferenciação Celular , Camundongos , Osteoclastos/fisiologia , Ligante RANK/fisiologia
15.
Aging (Albany NY) ; 12(8): 7030-7041, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32312943

RESUMO

Heterotopic ossification (HO) is a poorly characterized disease with ectopic bone formation in the musculoskeletal soft tissues. HO is widely considered as a tissue repair process goes away, with endochondral ossification to be the major pathological basis. The molecular mechanism of how the resident/recruited progenitor cells for tissue regeneration error differentiated into the chondrocytes remains unknown. Here, we found Transforming Growth Factor B Induced Gene Human Clone 3 (ßig-h3) was highly expressed in the inflammation and chondrogenesis stages of a heterotopic ossification model after rat Achilles tendon injury, as well as upon chondrogenic differentiation conditions in vitro. ßig-h3 functioned as an extracellular matrix protein, which was induced by TGFß signaling, could bind to the injured tendon-derived stem cells (iTDSCs) and inhibit the attachment of iTDSCs to collagen I. Exogenous ßig-h3 was also found able to accelerate the process of mesenchymal condensation of cultured iTDSCs and promote chondrogenic differentiation in vitro, and additional injection of iTDSCs could promote endochondral ossification in Achilles tendon injury model. Taken together, ßig-h3 might function as an adhesion protein that inhibited the attachment of iTDSCs to collagen I (the injury site) but promoted the attachment of iTDSCs to each other, which resulted in promoting chondrogenic differentiation.


Assuntos
Tendão do Calcâneo/patologia , Condrogênese/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Células-Tronco Mesenquimais/citologia , Ossificação Heterotópica/fisiopatologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Adesão Celular , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Feminino , Células-Tronco Mesenquimais/metabolismo , Ossificação Heterotópica/patologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Fator de Crescimento Transformador beta/análise
16.
Med Sci Monit ; 26: e918099, 2020 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-31915327

RESUMO

Fibulin-2, an extracellular matrix (ECM) protein expressed in normal epithelia, is a kind of fibulin which is associated with basement membranes (BM) and elastic ECM fibers. The role of fibulin-2 has been recognized as an oncogene. The upregulation of fibulin-2 correlates with cancer development and progression. Furthermore, the upregulation of fibulin has been detected in ovarian cancer and stomach adenocarcinoma. However, the downregulation of fibulin has been detected in different intestinal and respiratory tumor cells. Additional studies have revealed that the role of fibulin-2 in carcinogenesis is context dependent and is caused by the interaction of fibulin proteins such as cell surface receptors and other ECM proteins, including integrins and syndecans. The present study summarizes the role of fibulin in carcinogenesis and its underlying molecular mechanism.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Carcinogênese/metabolismo , Proteínas da Matriz Extracelular/fisiologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Feminino , Humanos , Integrinas/metabolismo , Neoplasias Ovarianas/metabolismo
17.
J Dent Res ; 98(10): 1066-1072, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31276626

RESUMO

Cells have been identified in postnatal tissues that, when isolated from multiple mesenchymal compartments, can be stimulated in vitro to give rise to cells that resemble mature mesenchymal phenotypes, such as odontoblasts, osteoblasts, adipocytes, and myoblasts. This has made these adult cells, collectively called mesenchymal stem cells (MSCs), strong candidates for fields such as tissue engineering and regenerative medicine. Based on evidence from in vivo genetic lineage-tracing studies, pericytes have been identified as a source of MSC precursors in vivo in multiple organs, in response to injury or during homeostasis. Questions of intense debate and interest in the field of tissue engineering and regenerative studies include the following: 1) Are all pericytes, irrespective of tissue of isolation, equal in their differentiation potential? 2) What are the mechanisms that regulate the differentiation of MSCs? To gain a better understanding of the latter, recent work has utilized ChIP-seq (chromatin immunoprecipitation followed by sequencing) to reconstruct histone landscapes. This indicated that for dental pulp pericytes, the odontoblast-specific gene Dspp was found in a transcriptionally permissive state, while in bone marrow pericytes, the osteoblast-specific gene Runx2 was primed for expression. RNA sequencing has also been utilized to further characterize the 2 pericyte populations, and results highlighted that dental pulp pericytes are already precommitted to an odontoblast fate based on enrichment analysis indicating overrepresentation of key odontogenic genes. Furthermore, ChIP-seq analysis of the polycomb repressive complex 1 component RING1B indicated that this complex is likely to be involved in inhibiting inappropriate differentiation, as it localized to a number of loci of key transcription factors that are needed for the induction of adipogenesis, chondrogenesis, or myogenesis. In this review, we highlight recent data elucidating molecular mechanisms that indicate that pericytes can be tissue-specific precommitted MSC precursors in vivo and that this precommitment is a major driving force behind MSC differentiation.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Pericitos/citologia , Adipogenia , Condrogênese , Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Humanos , Desenvolvimento Muscular , Fosfoproteínas/fisiologia , Complexo Repressor Polycomb 1/fisiologia , Sialoglicoproteínas/fisiologia , Fatores de Transcrição/fisiologia
18.
Clin Exp Metastasis ; 36(4): 351-363, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31119444

RESUMO

The infiltration of tumor-associated macrophages (TAMs) is associated with tumor progression and poor prognosis in endometrial cancer (EC). Collagen triple helix repeat containing 1 (CTHRC1), a secreted ECM protein, has been reported to have important roles in promoting cancer invasion and metastasis, but the functional role of CTHRC1 and its association with TAMs in EC remain unclear. Here we report that, in EC patients, CTHRC1 expression was up-regulated in endometrial cancer tissues compared with normal endometrium (P < 0.0001), and is positively correlated with tumor grade and depth of myometrial invasion (P = 0.024 and P = 0.0002, respectively). Meanwhile, CTHRC1 expression was positively correlated with an increased number of infiltrating TAMs, especially M2-like TAMs (P = 0.003, P = 0.001). In the tumor microenvironment of EC, CTHRC1 not only promoted myometrial invasion by interacting with Integrin ß3-Akt signaling pathway, but also promoted infiltration of M2-like TAMs by upregulating Fractalkine chemokine receptor (CX3CR1) expression in macrophages. Changing levels of recombinant CTHRC1 protein (rCTHRC1) promoted tumor migration and invasion via enhancing macrophage recruitment in vitro. In summary, our findings eventually provided a novel role for CTHRC1 in remodeling the tumor immune microenvironment to promote tumor metastasis in EC patients.


Assuntos
Neoplasias do Endométrio/patologia , Proteínas da Matriz Extracelular/fisiologia , Integrina beta3/fisiologia , Macrófagos/fisiologia , Miométrio/patologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Antígenos CD/análise , Antígenos de Diferenciação Mielomonocítica/análise , Linhagem Celular Tumoral , Movimento Celular , Neoplasias do Endométrio/imunologia , Feminino , Humanos , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/fisiologia , Receptores de Superfície Celular/análise , Transdução de Sinais/fisiologia , Microambiente Tumoral
19.
Endocrinology ; 160(3): 684-698, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30715257

RESUMO

Placental development is particularly altered in trisomy of chromosome 21 (T21)-affected pregnancies. We previously described in T21-affected placentae an abnormal paracrine crosstalk between the villus mesenchymal core and villus trophoblasts. T21-affected placentae are known to be characterized by their hypovascularity. However, the causes of this anomaly remain not fully elucidated. Therefore, the hypothesis of an abnormal paracrine crosstalk between fetal mesenchymal core and placental endothelial cells (PLECs) was evocated. Villus mesenchymal cells from control (CMCs) and T21 placentae (T21MCs) were isolated and grown in culture to allow their characterization and collection of conditioned media for functional analyses (CMC-CM and T21MC-CM, respectively). Interestingly, PLEC proliferation and branching ability were less stimulated by T21MC-CM than by CMC-CM. Protein array analysis identified secreted proangiogenic growth factors in CMC-CM, which were reduced in T21MC-CM. Combined mass spectrometry and biochemical analysis identified spondin-2 as a factor decreased in T21MC-CM compared with CMC-CM. We found that exogenous spondin-2 stimulated PLEC proliferation and established that T21MC-CM supplemented with spondin-2 recovered conditioned media ability to induce PLEC proliferation and angiogenesis. Hence, this study demonstrates a crosstalk between villus mesenchymal and fetal endothelial cells, in which spondin-2 secreted from mesenchymal cells plays a central role in placental vascular functions. Furthermore, our results also suggest that a reduction in spondin-2 secretion may contribute to the pathogenesis of T21 placental hypovascularity.


Assuntos
Síndrome de Down/fisiopatologia , Proteínas da Matriz Extracelular/fisiologia , Proteínas de Neoplasias/fisiologia , Placenta/irrigação sanguínea , Placentação , Estudos de Casos e Controles , Endostatinas/metabolismo , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neovascularização Fisiológica , Placenta/fisiopatologia , Gravidez
20.
Mol Carcinog ; 58(5): 621-626, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30582228

RESUMO

Loss-of-function of RHAMM causes hypofertility and testicular atrophy in young mice, followed by germ cell neoplasia in situ (GCNIS) of the testis, cellular atypia, and development of the testicular germ cell tumor (TGCT) seminoma. These pathologies reflect the risk factors and phenotypes that precede seminoma development in humans and-given the high prevalence of RHAMM downregulation in human seminoma-link RHAMM dysfunction with the aetiology of male hypofertility and GCNIS-related TGCTs. The initiating event underlying these pathologies, in RHAMM mutant testis, is premature displacement of undifferentiated progenitors from the basal compartment. We hypothesized that cd44 (both cancer initiating cell- and oncogenic progression marker) will drive GCNIS development, induced by RHAMM-loss-of-function in the mouse. We report that cd44 is expressed in a specific subset of GCNIS testes. Its genetic deletion has no effect on GCNIS onset, but it ameliorates oncogenic progression. We conclude that cd44 expression, combined with RHAMM dysfunction, promotes oncogenic progression in the testis.


Assuntos
Carcinoma in Situ/prevenção & controle , Proteínas da Matriz Extracelular/fisiologia , Receptores de Hialuronatos/fisiologia , Infertilidade Masculina/prevenção & controle , Neoplasias Embrionárias de Células Germinativas/prevenção & controle , Lesões Pré-Cancerosas/prevenção & controle , Neoplasias Testiculares/prevenção & controle , Animais , Biomarcadores Tumorais/genética , Carcinoma in Situ/genética , Carcinoma in Situ/metabolismo , Feminino , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/metabolismo , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/metabolismo , Deleção de Sequência , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA