Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Future Oncol ; 16(31): 2471-2474, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32772563

RESUMO

Over a century of clinical practice has led to the accumulation of millions of archived formalin fixed paraffin embedded (FFPE) cancer specimens with detailed medical records worldwide. Absolute quantitation of clinical protein biomarkers in these FFPE specimens allows individual specimens to be profiled at the population level, with the absolute nature of the measurements enabling the continuous processing of archived FFPE specimens over the time. A continuously growing cancer patient profile database is proposed here to support "big data" profiling of these protein biomarkers alone or in combination, enabling next-generation retrospective-prospective analytics into the field of clinical diagnostics.


Assuntos
Biomarcadores , Bases de Dados de Proteínas , Neoplasias/diagnóstico , Neoplasias/metabolismo , Proteoma , Proteômica , Técnicas Citológicas , Humanos , Neoplasias/etiologia , Inclusão em Parafina , Proteômica/métodos , Fixação de Tecidos
2.
Connect Tissue Res ; 58(2): 155-161, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27267924

RESUMO

Establishing a definitive diagnosis between benign enchondroma versus low-grade chondrosarcoma presents a potential challenge to both clinicians and pathologists. microRNAs (small non-coding RNAs) have proven to be effective biomarkers for the identification of tumors and tumor progression. We present analysis, both array and quantitative PCR, that shows consistently and substantially increased expression of two microRNAs, miRs-181a and -138, in low-grade chondrosarcomas compared with enchondromas. The data suggest these microRNAs would provide an analytical distinction between the chondrosarcoma and benign neoplasms that can be performed in formalin-fixed paraffin-embedded specimens. Together with recent publications, these data indicate that miRs-181a and -138 also play a role in tumor development and homeostasis and may provide new targets for the development of much needed therapeutic intervention.


Assuntos
Biomarcadores Tumorais , Condroma , Condrossarcoma , MicroRNAs , RNA Neoplásico , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Condroma/diagnóstico , Condroma/genética , Condroma/metabolismo , Condroma/patologia , Condrossarcoma/diagnóstico , Condrossarcoma/genética , Condrossarcoma/metabolismo , Condrossarcoma/patologia , Feminino , Humanos , Masculino , MicroRNAs/biossíntese , MicroRNAs/genética , Pessoa de Meia-Idade , RNA Neoplásico/biossíntese , RNA Neoplásico/genética
3.
Connect Tissue Res ; 56(3): 236-43, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25738598

RESUMO

AIM: The purpose of this study was to identify the microRNAs that regulate the response of chondrocytes to loss of matrix interaction. MATERIALS AND METHODS: MicroRNA and gene expression was compared in bovine cartilage and isolated chondrocytes using array analysis. Those microRNAs showing more than three-fold change in expression were verified by quantitative PCR after a stem-loop reverse transcription in bovine and human cartilage, and chondrocytes. Their function was investigated using target gene reporter construct expression, quantification of cell proliferation, and analysis of gene expression and matrix synthesis after transfection with microRNA mimics. RESULTS: Only four microRNAs were confirmed to have a greater than three-fold change in expression after isolation of bovine or human chondrocytes from their extracellular matrix; miRs-221, -222 and -21 showed increased expression and miR-483-5p showed decreased expression. Transfection with a miR-221 mimic was shown to suppress expression of the cyclin-dependent kinase inhibitor p27 leading to the stimulation of chondrocyte proliferation. Transfection of chondrocytes with a miR-483-5p mimic was shown to suppress several members of the mitogen activated protein kinase (MAPK) pathway; a likely explanation of the increased matrix production observed. CONCLUSIONS: microRNAs 221 and 483-5p respond to the loss of chondrocyte matrix interaction by respectively stimulating proliferation by suppression of inhibitors of cell division and suppression of matrix production possibly by release of inhibition of the MAPK pathway.


Assuntos
Proliferação de Células/genética , Condrócitos/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Expressão Gênica/genética , MicroRNAs/genética , Animais , Cartilagem Articular/metabolismo , Bovinos , Humanos , Transdução de Sinais/fisiologia
4.
Res Sq ; 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38464061

RESUMO

Vascular fibrosis, characterized by increased Type I collagen expression, significantly contributes to vascular remodeling. Our previous studies show that disrupting the expression of SM22α (aka SM22, Tagln) induces extensive vascular remodeling following arterial injury, involving oxidative stress, inflammation, and chondrogenesis within the vessel wall. This study aims to investigate the molecular mechanisms underlying the transcription of Col1a2, a key fibrotic extracellular matrix marker. We observed upregulation of COL1A2 in the arterial wall of Sm22-/- mice following carotid injury. Bioinformatics and molecular analyses reveal that Col1a2 transcription depends on a CArG box in the promoter, activated synergistically by SRF and SMAD3. Notably, we detected enhanced nuclear translocation of both SRF and SMAD3 in the smooth muscle cells of the injured carotid artery in Sm22-/- mice. These findings demonstrate that SM22 deficiency regulates vascular fibrosis through the interaction of SRF and the SMAD3-mediated canonical TGF-ß1 signal pathway, suggesting SM22α as a potential therapeutic target for preventing vascular fibrosis.

5.
Front Oncol ; 13: 920698, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36969021

RESUMO

Inherent issues of subjectivity and inconsistency have long plagued immunohistochemistry (IHC)-based Her2 assessment, leading to the repeated issuance of guidelines by the American Society of Clinical Oncology/College of American Pathologists (ASCO/CAP) for its standardization for breast cancer patients. Yet, all these efforts may prove insufficient with the advent of Trastuzumab deruxtecan (T-Dxd), a drug with the promise to expand to tumors traditionally defined as Her2 negative (Her2-). In this study, we attempted to address these issues by exploring an ELISA-like quantitative dot blot (QDB) method as an alternative to IHC. The QDB method has been used to measure multiple protein biomarkers including ER, PR, Ki67, and cyclin D1 in breast cancer specimens. Using an independent cohort (cohort 2) of breast cancer formalin-fixed paraffin-embedded (FFPE) specimens, we validated cutoffs developed in cohort 1 (Yu et al., Scientific Reports 2020 10:10502) with overall 100% specificity (95% CI: 100-100) and 97.56% sensitivity (95% CI: 92.68-100) in cohort 2 against standard practice with the dichotomized absolutely quantitated values. Using the limit of detection (LOD) of the QDB method as the putative cutoff point, tumors with no Her2 expression were identified with the number comparable to those of IHC 0. Our results support further evaluation of the QDB method as an alternative to IHC to meet the emerging need of identifying tumors with low Her2 expression (Her2-low) in daily clinical practice.

6.
Circ Res ; 106(8): 1351-62, 2010 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-20224039

RESUMO

RATIONALE: SM22 (or transgelin), an actin-binding protein abundant in vascular smooth muscle cells (VSMCs), is downregulated in atherosclerosis, aneurysm and various cancers. Abolishing SM22 in apolipoprotein E knockout mice accelerates atherogenesis. However, it is unclear whether SM22 disruption independently promotes arterial inflammation. OBJECTIVE: To investigate whether SM22 disruption directly promotes inflammation on arterial injury and to characterize the underlying mechanisms. METHODS AND RESULTS: Using carotid denudation as an artery injury model, we showed that Sm22 knockout (Sm22(-/-)) mice developed enhanced inflammatory responses with higher induction of proinflammatory genes, including Vcam1, Icam1, Cx3cl1, Ccl2, and Ptgs2. Higher expression of these genes was confirmed in primary Sm22(-/-) VSMCs and in PAC1 cells after Sm22 knockdown, whereas SM22 recapitulation in primary Sm22(-/-) VSMCs decreased their expression. NFKB2 was prominently activated in both injured carotids of Sm22(-/-) mice and in PAC1 cells after Sm22 knockdown and may mediate upregulation of these proinflammatory genes. As a NF-kappaB activator, reactive oxygen species (ROS) increased in primary Sm22(-/-) VSMCs and in PAC1 cells after Sm22 knockdown. ROS scavengers blocked NF-kappaB activation and induction of proinflammatory genes. Furthermore, Sm22 knockdown increased Sod2 expression and activated p47phox, reflecting contributions of mitochondria and NADPH oxidase to the augmented ROS production; this may result from actin and microtubule cytoskeletal remodeling. CONCLUSIONS: Our findings show that SM22 downregulation can induce proinflammatory VSMCs through activation of ROS-mediated NF-kappaB pathways. This study provides initial evidence linking VSMC cytoskeleton remodeling with arterial inflammation.


Assuntos
Lesões das Artérias Carótidas/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Animais , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Células Cultivadas , Citoesqueleto/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Inflamação/genética , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Mitocôndrias Musculares/metabolismo , Proteínas Musculares/deficiência , Proteínas Musculares/genética , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , NADPH Oxidases/metabolismo , Subunidade p52 de NF-kappa B/metabolismo , Oxirredução , Interferência de RNA , RNA Mensageiro/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/genética , Superóxido Dismutase/metabolismo , Fator de Transcrição RelA/metabolismo
7.
J Gastric Cancer ; 21(4): 335-351, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35079437

RESUMO

PURPOSE: An underlying factor for the failure of several clinical trials of anti-epidermal growth factor receptor (EGFR) therapies is the lack of an effective method to identify patients who overexpress EGFR protein. The quantitative dot blot method (QDB) was used to measure EGFR protein levels objectively, absolutely, and quantitatively. Its feasibility was evaluated for the prognosis of overall survival (OS) of patients with gastric cancer. MATERIALS AND METHODS: Slices of 2×5 µm from formalin-fixed paraffin-embedded gastric cancer specimens were used to extract total tissue lysates for QDB measurement. Absolutely quantitated EGFR protein levels were used for the Kaplan-Meier OS analysis. RESULTS: EGFR protein levels ranged from 0 to 772.6 pmol/g (n=246) for all gastric cancer patients. A poor correlation was observed between quantitated EGFR levels and immunohistochemistry scores with ρ=0.024 and P=0.717 in Spearman's correlation analysis. EGFR was identified as an independent negative prognostic biomarker for gastric cancer patients only through absolute quantitation, with a hazard ratio of 1.92 (95% confidence interval, 1.05-3.53; P=0.034) in multivariate Cox regression OS analysis. A cutoff of 208 pmol/g was proposed to stratify patients with a 3-year survival probability of 44% for patients with EGFR levels above the cutoff versus 68% for those below the cutoff based on Kaplan-Meier OS analysis (log rank test, P=0.002). CONCLUSIONS: A QDB-based assay was developed for gastric cancer specimens to measure EGFR protein levels absolutely, quantitatively, and objectively. This assay should facilitate clinical trials aimed at evaluation of anti-EGFR therapies retrospectively and prospectively for gastric cancer.

8.
Connect Tissue Res ; 51(3): 230-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20073988

RESUMO

Matrix metalloproteinases (MMP)-13 activity is necessary for normal skeletal development and plays a central role in cartilage degeneration associated with osteoarthritis (OA). The studies we described here examine the interactions of the hemopexin domain of MMP-13 with proteins secreted by human chondrocytes in culture. The hemopexin domain of the MMPs and many other proteins in which this structure is found mediates protein function by forming the primary site of interaction with other proteins. We have modified a tandem affinity expression tag (hTAP) to enable efficient expression of the tagged bait protein. In this case the MMP-13 C-terminal domain (CTD) comprises hinge and hemopexin domain, and we immobilized the fusion construct on a column of agarose bound immunoglobin G. The MMP-13 CTD affinity column so generated enabled the efficient and gentle isolation of interacting proteins from the culture medium of human articular chondrocytes. TIMP1 and alpha2-macroglobulin previously shown to interact with MMP-13 as well as several proteins, fibronectin, type VI collagen and xylosyltransferase 1 and several proteoglycans, decorin, syndecan 4 and serglycin not previously recognized as interacting with MMP-13 were identified by mass spectrometry. The interaction between isolated proteins and MMP-13 CTD was verified by yeast two hybrid analysis. We also demonstrated serglycin expression by chondrocytes for the first time and its co localization with MMP-13 in a cytoplasmic granular morphology. The consequence of these interactions remains to be demonstrated, however; binding to MMP-13 suggests a role in the regulation of cartilage degradation.


Assuntos
Domínio Catalítico , Condrócitos/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Células Cultivadas , Condrócitos/patologia , Cromatografia de Afinidade , Clonagem Molecular , Sequência Conservada , Hemopexina/química , Hemopexina/metabolismo , Rim , Espectrometria de Massas , Metaloproteinase 13 da Matriz/química , Osteoartrite/patologia , Mapeamento de Peptídeos , Ligação Proteica , Proteoglicanas/metabolismo , Proteínas de Transporte Vesicular/metabolismo
9.
Exp Mol Pathol ; 88(1): 52-7, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19874818

RESUMO

Aorta organ culture has been widely used as an ex vivo model for studying vessel pathophysiology. Recent studies show that the vascular smooth muscle cells (VSMCs) in organ culture undergo drastic dedifferentiation within the first few hours (termed early phenotypic modulation). Loss of tensile stress to which aorta is subject in vivo is the cause of this early phenotypic modulation. However, no underlying molecular mechanism has been discovered thus far. The purpose of the present study is to identify intracellular signals involved in the early phenotypic modulation of VSMC in organ culture. We find that the drastic VSMC dedifferentiation is accompanied by accelerated actin cytoskeleton dynamics and downregulation of SRF and myocardin. Among the variety of signal pathways examined, increasing actin polymerization by jasplakinolide is the only one hindering VSMC dedifferentiation in organ culture. Moreover, jasplakinolide reverses actin dynamics during organ culture. Latrunculin B (disrupting actin cytoskeleton) and jasplakinolide respectively suppressed and enhanced the expression of VSMC markers, SRF, myocardin, and CArG-box-mediated SMC promoters in PAC1, a VSMC line. These results identify actin cytoskeleton degradation as a major intracellular signal for loss of tensile stress-induced early phenotypic modulation of VSMC in organ culture. This study suggests that disrupting actin cytoskeleton integrity may contribute to the pathogenesis of vascular diseases.


Assuntos
Actinas/fisiologia , Desdiferenciação Celular/genética , Citoesqueleto/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Animais , Biomarcadores/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Desdiferenciação Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Depsipeptídeos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Técnicas de Cultura de Órgãos , Fenótipo , Estresse Mecânico , Tiazolidinas/farmacologia , Transativadores/genética , Transativadores/metabolismo , Proteínas Elk-4 do Domínio ets/genética , Proteínas Elk-4 do Domínio ets/metabolismo
10.
Sci Rep ; 10(1): 21781, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33311559

RESUMO

Yin Yang 1 (YY1) regulates gene transcription in a variety of biological processes. In this study, we aim to determine the role of YY1 in vascular smooth muscle cell (VSMC) phenotypic modulation both in vivo and in vitro. Here we show that vascular injury in rodent carotid arteries induces YY1 expression along with reduced expression of smooth muscle differentiation markers in the carotids. Consistent with this finding, YY1 expression is induced in differentiated VSMCs in response to serum stimulation. To determine the underlying molecular mechanisms, we found that YY1 suppresses the transcription of CArG box-dependent SMC-specific genes including SM22α, SMα-actin and SMMHC. Interestingly, YY1 suppresses the transcriptional activity of the SM22α promoter by hindering the binding of serum response factor (SRF) to the proximal CArG box. YY1 also suppresses the transcription and the transactivation of myocardin (MYOCD), a master regulator for SMC-specific gene transcription by binding to SRF to form the MYOCD/SRF/CArG box triad (known as the ternary complex). Mechanistically, YY1 directly interacts with MYOCD to competitively displace MYOCD from SRF. This is the first evidence showing that YY1 inhibits SMC differentiation by directly targeting MYOCD. These findings provide new mechanistic insights into the regulatory mechanisms that govern SMC phenotypic modulation in the pathogenesis of vascular diseases.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fator de Resposta Sérica/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Fator de Transcrição YY1/metabolismo , Animais , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley
11.
J Chromatogr B Analyt Technol Biomed Life Sci ; 875(2): 493-501, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18952512

RESUMO

Like other members of the transforming growth factor beta (TGF-beta) family of growth factors, the biological activity of TGF-beta2 is believed to be regulated by the formation and dissociation of multiprotein complexes. To isolate the molecular complex formed by TGF-beta2 secreted by hypertrophic chondrocytes we have used expression of TGF-beta2 fused with the humanized, tandem affinity purification (hTAP) tag and mass spectrometry for the identification of interacting proteins. The hTAP synthetic gene was assembled by systematically replacing the rare codons of the original TAP tag with codons most preferred in highly expressed human genes to circumvent the poor translation efficiency of the original TAP tag in animal cells. TGF-beta2 was shown to interact with Type X collagen and this interaction confirmed using V5 tagged TGF-beta2. Functional interaction was suggested by the inhibition of TGF-beta2 activity by type X collagen in culture and the influence of a mutation in type X collagen on the distribution of TGF-beta2 in growth cartilage.


Assuntos
Clonagem Molecular , Colágeno Tipo X/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Adolescente , Marcadores de Afinidade/metabolismo , Animais , Células Cultivadas , Galinhas , Criança , Condrócitos/metabolismo , Colágeno Tipo X/genética , Fibroblastos/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Mutação , Oligopeptídeos , Osteocondrodisplasias/patologia , Peptídeos/genética , Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/metabolismo , Espectrometria de Massas por Ionização por Electrospray , Transfecção , Fator de Crescimento Transformador beta2/genética
12.
Am J Clin Pathol ; 128(3): 382-8, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17709311

RESUMO

The nuclear immunoreactivity for thyroid transcription factor-1 (TTF-1) is a useful marker for identification of carcinomas of thyroid and lung origin. Our aim was to determine whether cytoplasmic staining in the liver is a result of cross-reaction of anti-TTF-1 antibody (clone 8G7G3/1, DAKO, Carpinteria, CA) or true positivity resulting from aberrant expression of TTF-1 or products of the alternatively sliced TTF-1 gene. Fresh tissue samples from liver, thyroid, and lung were obtained for H&E-stained sections, TTF-1 immunostaining, and RNA and protein analyses. Western blot revealed an abundant band corresponding to an approximately 160-kd protein from liver but not either thyroid or lung tissue samples. By reverse transcriptase-polymerase chain reaction, messenger RNA of TTF-1 was not detectable in liver tissue. Our study demonstrates that TTF-1 immunoreactivity (clone 8G7G3/1) in the hepatocyte cytoplasm is due to an approximately 160-kd protein; this unique protein is not an alternative splicing product of TTF-1 and neither is it expressed in thyroid and lung tissues.


Assuntos
Citoplasma/química , Hepatócitos/química , Proteínas Nucleares/análise , Fatores de Transcrição/análise , Processamento Alternativo , Anticorpos/metabolismo , Reações Cruzadas , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/metabolismo , Proteínas Nucleares/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Glândula Tireoide/química , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/imunologia
13.
Nat Rev Rheumatol ; 13(4): 205-216, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28202911

RESUMO

CRISPR/Cas9 genome editing technology has taken the research world by storm since its use in eukaryotes was first proposed in 2012. Publications describing advances in technology and new applications have continued at an unrelenting pace since that time. In this Review, we discuss the application of CRISPR/Cas9 for creating gene mutations - the application that initiated the current avalanche of interest - and new developments that have largely answered initial concerns about its specificity and ability to introduce new gene sequences. We discuss the new, diverse and rapidly growing adaptations of the CRISPR/Cas9 technique that enable activation, repression, multiplexing and gene screening. These developments have enabled researchers to create sophisticated tools for dissecting the function and inter-relatedness of genes, as well as noncoding regions of the genome, and to identify gene networks and noncoding regions that promote disease or confer disease susceptibility. These approaches are beginning to be used to interrogate complex and multilayered biological systems and to produce complex animal models of disease. CRISPR/Cas9 technology has enabled the application of new therapeutic approaches to treating disease in animal models, some of which are beginning to be seen in the first human clinical trials. We discuss the direct application of these techniques to rheumatic diseases, which are currently limited but are sure to increase rapidly in the near future.


Assuntos
Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Edição de Genes/métodos , Terapia Genética/métodos , RNA Guia de Cinetoplastídeos , Doenças Reumáticas/genética , Animais , Regulação da Expressão Gênica , Testes Genéticos , Humanos , Mutação , Doenças Reumáticas/terapia
14.
PLoS One ; 12(12): e0190191, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29284006

RESUMO

Vascular smooth muscle cell (VSMC) phenotypic modulation is characterized by the downregulation of SMC actin cytoskeleton proteins. Our published study shows that depletion of SM22α (aka SM22, Transgelin, an actin cytoskeleton binding protein) promotes inflammation in SMCs by activating NF-κB signal pathways both in cultured VSMCs and in response to vascular injury. The goal of this study is to investigate the underlying molecular mechanisms whereby SM22 suppresses NF-κB signaling pathways under inflammatory condition. NF-κB inducing kinase (Nik, aka MAP3K14, activated by the LTßR) is a key upstream regulator of NF-κB signal pathways. Here, we show that SM22 overexpression suppresses the expression of NIK and its downstream NF-κB canonical and noncanonical signal pathways in a VSMC line treated with a LTßR agonist. SM22 regulates NIK expression at both transcriptional and the proteasome-mediated post-translational levels in VSMCs depending on the culture condition. By qPCR, chromatin immunoprecipitation and luciferase assays, we found that Nik is a transcription target of serum response factor (SRF). Although SM22 is known to be expressed in the cytoplasm, we found that SM22 is also expressed in the nucleus where SM22 interacts with SRF to inhibit the transcription of Nik and prototypical SRF regulated genes including c-fos and Egr3. Moreover, carotid injury increases NIK expression in Sm22-/- mice, which is partially relieved by adenovirally transduced SM22. These findings reveal for the first time that SM22 is expressed in the nucleus in addition to the cytoplasm of VSMCs to regulate the transcription of Nik and its downstream proinflammatory NF-kB signal pathways as a modulator of SRF during vascular inflammation.


Assuntos
Citocinas/fisiologia , Inflamação/fisiopatologia , Proteínas dos Microfilamentos/fisiologia , Proteínas Musculares/fisiologia , Músculo Liso Vascular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transcrição Gênica , Animais , Linhagem Celular , Camundongos , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética , Músculo Liso Vascular/citologia , Proteínas Serina-Treonina Quinases/genética , Quinase Induzida por NF-kappaB
16.
Sci Rep ; 5: 10180, 2015 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-25985281

RESUMO

TGF-ß signaling plays critical roles in the pathogenesis of aneurysms; however, it is still unclear whether its role is protective or destructive. In this study, we investigate the role of SMAD3 in the pathogenesis of calcium chloride (CaCl2)-induced abdominal aortic aneurysms (AAA) in Smad3(-/-), Smad3(+/-) and Smad3(+/+) mice. We find that loss of SMAD3 drastically increases wall thickening of the abdominal aorta. Histological analyses show significant vessel wall remodeling with elastic fiber fragmentation. Remarkably, under polarized light, collagen fibers in the hyperplastic adventitia of Smad3(-/-) mice show extensive reorganization accompanied by loosely packed thin and radial collagen fibers. The expressions of matrix metalloproteinases including MMP2, MMP9, and MMP12 and infiltration of macrophage/T cells are drastically enhanced in the vascular wall of Smad3(-/-) mice. We also observe marked increase of NF-κB and ERK1/2 signaling as well as the expression of nuclear Smad2, Smad4 and TGF-ß1 in the vessel wall of Smad3(-/-) mice. In addition, we find that SMAD3 expression is reduced in the dedifferentiated medial smooth muscle-like cells of human AAA patients. These findings provide direct in vivo evidence to support the essential roles of SMAD3 in protecting vessel wall integrity and suppressing inflammation in the pathogenesis of AAAs.


Assuntos
Aneurisma da Aorta Abdominal/etiologia , Remodelamento Atrial/genética , Colágeno/metabolismo , Inflamação/complicações , Leucócitos/metabolismo , Proteína Smad3/deficiência , Animais , Aneurisma da Aorta Abdominal/patologia , Cloreto de Cálcio/farmacologia , Modelos Animais de Doenças , Elastina , Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Leucócitos/patologia , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Knockout , NF-kappa B , Transdução de Sinais , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/metabolismo
17.
Brain Res ; 1030(1): 19-27, 2004 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-15567334

RESUMO

Human bone marrow stromal cells (hMSCs) enhance neurological recovery after stroke in rodents, possibly via induction of growth factors. We therefore elected to test the effects of hMSC treatment on insulin-like growth factor 1 (IGF-1), which plays an important role in growth, development, neuroprotection and repair in the adult. Rats (n=57) were subjected to permanent middle cerebral artery occlusion (MCAo) and injected intravenously with 3 x 10(6) hMSCs or phosphate-buffered saline (PBS) at 1 day after MCAo. Functional outcome was measured after MCAo using a modified Neurological Severity Score (mNSS). Gene expression of IGF-1 and IGF-1 receptor (IGF-1R) in the ischemic brain tissue were measured at 2 and 7 days after MCAo using reverse transcription-polymerase chain reaction (RT-PCR). Immunohistochemistry was performed to measure the expression of bromodeoxyuridine (BrdU), doublecortin (DCX), IGF-1 and IGF-1R at 7, 14 and 30 days after MCAo. Treatment of MCAo with hMSCs significantly improved functional recovery from 14 to 30 days. MAB1281-labeled hMSCs entered the ischemic brain and increased time-dependently. hMSC treatment significantly increased IGF-1 mRNA and BrdU(+), DCX(+), IGF-1(+) and IGF-1R(+) cells compared to PBS-treated rats (p<0.05). The percentage of BrdU(+) or DCX(+) cells colocalized with IGF-1 increased in the hMSC-treated rats compared to the PBS-treated rats (p<0.05). IGF-1 and IGF-1R may contribute to improved functional recovery and increased neurogenesis after treatment of stroke with hMSCs.


Assuntos
Transplante de Medula Óssea , Isquemia Encefálica/terapia , Fator de Crescimento Insulin-Like I/genética , Receptor IGF Tipo 1/genética , Células Estromais/transplante , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Divisão Celular , Movimento Celular , Células Cultivadas , Proteína Duplacortina , Expressão Gênica , Humanos , Masculino , Neurônios/citologia , Neurônios/fisiologia , RNA Mensageiro/análise , Ratos , Ratos Wistar , Recuperação de Função Fisiológica , Células-Tronco/citologia , Células-Tronco/fisiologia , Acidente Vascular Cerebral/terapia , Células Estromais/citologia , Células Estromais/fisiologia , Transplante Heterólogo
18.
Bone ; 69: 118-25, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25260929

RESUMO

The Swarm rat chondrosarcoma (RCS) cell lines derived from a spontaneous neoplasm in a rat spine several decades ago have provided excellent models of chondrosarcoma tumor development. In addition the robust chondrocyte phenotype (expression of a large panel of genes identical to that seen in normal rat cartilage) and the ability to generate cell clones have facilitated their extensive use in the identification of chondrocyte proteins and genes. The clustered regularly interspersed short palindromic repeat (CRISPR) technology employing the RNA-guided nuclease Cas9 has rapidly dominated the genome engineering field as a unique and powerful gene editing tool. We have generated a stable RCS cell line (RCS Cas9) expressing the nuclease Cas9 that enables the editing of any target gene or non-coding RNA by simple transfection with a guide RNA. As proof of principle, stable cell lines with targeted ablation of aggrecan expression (Acan KO) were generated and characterized. The studies show that stable chondrocyte cell lines with targeted genome editing can be quickly generated from RCS Cas9 cells using this system. The Acan KO cell lines also provided a tool for characterizing the response of chondrocytes to aggrecan loss and the role of aggrecan in chondrosarcoma development. Loss of aggrecan expression while not affecting the chondrocyte phenotype resulted in a much firmer attachment of cells to their substrate in culture. Large changes in the expression of several genes were observed in response to the absence of the proteoglycan matrix, including those for several small leucine rich proteoglycans (SLRPs), transcription factors and membrane transporters. Acan KO cells failed to form a substantial chondrosarcoma when injected subcutaneously in nude mice consistent with previous suggestions that the glycosaminoglycan-rich matrix surrounding the chondrosarcoma protects it from destruction by the host immune system. The studies provide new understanding of aggrecan function and the RCS Cas9 cell line is expected to provide a very valuable tool for the study gene function in chondrocytes.


Assuntos
Agrecanas/genética , Condrócitos , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Técnicas de Inativação de Genes/métodos , Animais , Sequência de Bases , Western Blotting , Linhagem Celular , Desoxirribonucleases , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Fenótipo , Ratos , Ratos Sprague-Dawley , Transcriptoma , Transfecção
20.
Cardiovasc Res ; 90(1): 28-37, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21183509

RESUMO

AIMS: Expression of SM22 (also known as SM22alpha and transgelin), a vascular smooth muscle cells (VSMCs) marker, is down-regulated in arterial diseases involving medial osteochondrogenesis. We investigated the effect of SM22 deficiency in a mouse artery injury model to determine the role of SM22 in arterial chondrogenesis. METHODS AND RESULTS: Sm22 knockout (Sm22(-/-)) mice developed prominent medial chondrogenesis 2 weeks after carotid denudation as evidenced by the enhanced expression of chondrogenic markers including type II collagen, aggrecan, osteopontin, bone morphogenetic protein 2, and SRY-box containing gene 9 (SOX9). This was concomitant with suppression of VSMC key transcription factor myocardin and of VSMC markers such as SM α-actin and myosin heavy chain. The conversion tendency from myogenesis to chondrogenesis was also observed in primary Sm22(-/-) VSMCs and in a VSMC line after Sm22 knockdown: SM22 deficiency altered VSMC morphology with compromised stress fibre formation and increased actin dynamics. Meanwhile, the expression level of Sox9 mRNA was up-regulated while the mRNA levels of myocardin and VSMC markers were down-regulated, indicating a pro-chondrogenic transcriptional switch in SM22-deficient VSMCs. Furthermore, the increased expression of SOX9 was mediated by enhanced reactive oxygen species production and nuclear factor-κB pathway activation. CONCLUSION: These findings suggest that disruption of SM22 alters the actin cytoskeleton and promotes chondrogenic conversion of VSMCs.


Assuntos
Lesões das Artérias Carótidas/patologia , Transdiferenciação Celular , Condrócitos/patologia , Condrogênese , Proteínas dos Microfilamentos/deficiência , Proteínas Musculares/deficiência , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Túnica Média/patologia , Lesões do Sistema Vascular/patologia , Animais , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/metabolismo , Transdiferenciação Celular/genética , Células Cultivadas , Condrócitos/metabolismo , Condrogênese/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Genótipo , Masculino , Metaplasia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Desenvolvimento Muscular , Proteínas Musculares/genética , Músculo Liso Vascular/lesões , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , NF-kappa B/metabolismo , Oxirredução , Fenótipo , Interferência de RNA , RNA Mensageiro/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transfecção , Túnica Média/lesões , Túnica Média/metabolismo , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA