Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.142
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 158(2): 449-461, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25036638

RESUMEN

Reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) holds enormous promise for regenerative medicine. To elucidate endogenous barriers limiting this process, we systematically dissected human cellular reprogramming by combining a genome-wide RNAi screen, innovative computational methods, extensive single-hit validation, and mechanistic investigation of relevant pathways and networks. We identify reprogramming barriers, including genes involved in transcription, chromatin regulation, ubiquitination, dephosphorylation, vesicular transport, and cell adhesion. Specific a disintegrin and metalloproteinase (ADAM) proteins inhibit reprogramming, and the disintegrin domain of ADAM29 is necessary and sufficient for this function. Clathrin-mediated endocytosis can be targeted with small molecules and opposes reprogramming by positively regulating TGF-ß signaling. Genetic interaction studies of endocytosis or ubiquitination reveal that barrier pathways can act in linear, parallel, or feedforward loop architectures to antagonize reprogramming. These results provide a global view of barriers to human cellular reprogramming.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Proteínas ADAM/metabolismo , Adhesión Celular , Células Madre Embrionarias/metabolismo , Endocitosis , Humanos , Ubiquitina/metabolismo
2.
Cell ; 156(4): 730-43, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24529376

RESUMEN

Osteoarthritis (OA), primarily characterized by cartilage degeneration, is caused by an imbalance between anabolic and catabolic factors. Here, we investigated the role of zinc (Zn2+) homeostasis, Zn2+ transporters, and Zn(2+)-dependent transcription factors in OA pathogenesis. Among Zn2+ transporters, the Zn2+ importer ZIP8 was specifically upregulated in OA cartilage of humans and mice, resulting in increased levels of intracellular Zn2+ in chondrocytes. ZIP8-mediated Zn2+ influx upregulated the expression of matrix-degrading enzymes (MMP3, MMP9, MMP12, MMP13, and ADAMTS5) in chondrocytes. Ectopic expression of ZIP8 in mouse cartilage tissue caused OA cartilage destruction, whereas Zip8 knockout suppressed surgically induced OA pathogenesis, with concomitant modulation of Zn2+ influx and matrix-degrading enzymes. Furthermore, MTF1 was identified as an essential transcription factor in mediating Zn2+/ZIP8-induced catabolic factor expression, and genetic modulation of Mtf1 in mice altered OA pathogenesis. We propose that the zinc-ZIP8-MTF1 axis is an essential catabolic regulator of OA pathogenesis.


Asunto(s)
Osteoartritis/metabolismo , Osteoartritis/patología , Transducción de Señal , Proteínas ADAM/metabolismo , Anciano , Animales , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Condrocitos/metabolismo , Condrocitos/patología , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Regulación hacia Arriba , Zinc/metabolismo
3.
Mol Cell Proteomics ; 23(3): 100722, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38272115

RESUMEN

Glioblastoma (GBM) is the most aggressive brain tumor and different efforts have been employed in the search for new drugs and therapeutic protocols for GBM. Epitranscriptomics has shed light on new druggable Epigenetic therapies specifically designed to modulate GBM biology and behavior such as Histone Deacetylase inhibitors (iHDAC). Although the effects of iHDAC on GBM have been largely explored, there is a lack of information on the underlaying mechanisms HDAC-dependent that modulate the repertoire of GBM secreted molecules focusing on the set of Extracellular Matrix (ECM) associated proteins, the Matrisome, that may impact the surrounding tumor microenvironment. To acquire a better comprehension of the impacts of HDAC activity on the GBM Matrisome, we studied the alterations on the Matrisome-associated ECM regulators, Core Matrisome ECM glycoproteins, ECM-affiliated proteins and Proteoglycans upon HDAC inhibition in vitro as well as their relationship with glioma pathophysiological/clinical features and angiogenesis. For this, U87MG GBM cells were treated for with iHDAC or vehicle (control) and the whole secretome was processed by Mass Spectrometry NANOLC-MS/MS. In silico analyses revealed that proteins associated to the Angiogenic Matrisome (AngioMatrix), including Decorin, ADAM10, ADAM12 and ADAM15 were differentially regulated in iHDAC versus control secretome. Interestingly, genes coding for the Matrisome proteins differentially regulated were found mutated in patients and were correlated to glioma pathophysiological/clinical features. In vitro functional assays, using HBMEC endothelial cells exposed to the secretome of control or iHDAC treated GBM cells, coupled to 2D and 3D GBM cell culture system, showed impaired migratory capacity of endothelial cells and disrupted tubulogenesis in a Fibronectin and VEGF independent fashion. Collectively, our study provides understanding of epigenetic mechanisms HDAC-dependent to key Matrisomal proteins that may contribute to identify new druggable Epigenetic therapies or gliomagenesis biomarkers with relevant implications to improve therapeutic protocols for this malignancy.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Glioblastoma/genética , Glioblastoma/patología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Células Endoteliales/metabolismo , Espectrometría de Masas en Tándem , Matriz Extracelular/metabolismo , Glioma/metabolismo , Epigénesis Genética , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Microambiente Tumoral , Proteínas de la Membrana/metabolismo , Proteínas ADAM/metabolismo
4.
Rev Physiol Biochem Pharmacol ; 184: 69-120, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35061104

RESUMEN

Metalloproteinases are a group of proteinases that plays a substantial role in extracellular matrix remodeling and its molecular signaling. Among these metalloproteinases, ADAMs (a disintegrin and metalloproteinases) and ADAM-TSs (ADAMs with thrombospondin domains) have emerged as highly efficient contributors mediating proteolytic processing of various signaling molecules. ADAMs are transmembrane metalloenzymes that facilitate the extracellular domain shedding of membrane-anchored proteins, cytokines, growth factors, ligands, and their receptors and therefore modulate their biological functions. ADAM-TSs are secretory, and soluble extracellular proteinases that mediate the cleavage of non-fibrillar extracellular matrix proteins. ADAMs and ADAM-TSs possess pro-domain, metalloproteinase, disintegrin, and cysteine-rich domains in common, but ADAM-TSs have characteristic thrombospondin motifs instead of the transmembrane domain. Most ADAMs and ADAM-TSs are activated by cleavage of pro-domain via pro-protein convertases at their N-terminus, hence directing them to various signaling pathways. In this article, we are discussing not only the structure and regulation of ADAMs and ADAM-TSs, but also the importance of these metalloproteinases in various human pathophysiological conditions like cardiovascular diseases, colorectal cancer, autoinflammatory diseases (sepsis/rheumatoid arthritis), Alzheimer's disease, proliferative retinopathies, and infectious diseases. Therefore, based on the emerging role of ADAMs and ADAM-TSs in various human pathologies, as summarized in this review, these metalloproteases can be considered as critical therapeutic targets and diagnostic biomarkers.


Asunto(s)
Proteínas ADAM , Enfermedad , Desintegrinas , Humanos , Proteínas ADAM/química , Proteínas ADAM/metabolismo , Enfermedades Cardiovasculares/metabolismo , Trombospondinas , Biomarcadores/metabolismo
5.
Blood ; 141(24): 2993-3005, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37023370

RESUMEN

Antibody binding to a plasma metalloprotease, a disintegrin and metalloproteinase with thrombospondin type 1 repeats 13 (ADAMTS13), is necessary for the development of immune thrombotic thrombocytopenic purpura (iTTP). Inhibition of ADAMTS13-mediated von Willebrand factor (VWF) cleavage by such antibodies clearly plays a role in the pathophysiology of the disease, although the mechanisms by which they inhibit ADAMTS13 enzymatic function are not fully understood. At least some immunoglobulin G-type antibodies appear to affect the conformational accessibility of ADAMTS13 domains involved in both substrate recognition and inhibitory antibody binding. We used single-chain fragments of the variable region previously identified via phage display from patients with iTTP to explore the mechanisms of action of inhibitory human monoclonal antibodies. Using recombinant full-length ADAMTS13, truncated ADAMTS13 variants, and native ADAMTS13 in normal human plasma, we found that, regardless of the conditions tested, all 3 inhibitory monoclonal antibodies tested affected enzyme turnover rate much more than substrate recognition of VWF. Hydrogen-to-deuterium exchange plus mass spectrometry experiments with each of these inhibitory antibodies demonstrated that residues in the active site of the catalytic domain of ADAMTS13 are differentially exposed to solvent in the presence and absence of monoclonal antibody binding. These results support the hypothesis that inhibition of ADAMTS13 in iTTP may not necessarily occur because the antibodies directly prevent VWF binding, but instead because of allosteric effects that impair VWF cleavage, likely by affecting the conformation of the catalytic center in the protease domain of ADAMTS13. Our findings provide novel insight into the mechanism of autoantibody-mediated inhibition of ADAMTS13 and pathogenesis of iTTP.


Asunto(s)
Púrpura Trombocitopénica Idiopática , Púrpura Trombocitopénica Trombótica , Trombosis , Humanos , Anticuerpos Monoclonales , Factor de von Willebrand/metabolismo , Proteínas ADAM/química , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Autoanticuerpos
6.
PLoS Biol ; 20(1): e3001532, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35085231

RESUMEN

Chronic inflammation is often associated with the development of tissue fibrosis, but how mesenchymal cell responses dictate pathological fibrosis versus resolution and healing remains unclear. Defining stromal heterogeneity and identifying molecular circuits driving extracellular matrix deposition and remodeling stands to illuminate the relationship between inflammation, fibrosis, and healing. We performed single-cell RNA-sequencing of colon-derived stromal cells and identified distinct classes of fibroblasts with gene signatures that are differentially regulated by chronic inflammation, including IL-11-producing inflammatory fibroblasts. We further identify a transcriptional program associated with trans-differentiation of mucosa-associated fibroblasts and define a functional gene signature associated with matrix deposition and remodeling in the inflamed colon. Our analysis supports a critical role for the metalloprotease Adamdec1 at the interface between tissue remodeling and healing during colitis, demonstrating its requirement for colon epithelial integrity. These findings provide mechanistic insight into how inflammation perturbs stromal cell behaviors to drive fibroblastic responses controlling mucosal matrix remodeling and healing.


Asunto(s)
Proteínas ADAM/inmunología , Colitis/inmunología , Matriz Extracelular/metabolismo , Fibroblastos/inmunología , Mucosa Intestinal/inmunología , Células Madre Mesenquimatosas/inmunología , Proteínas ADAM/deficiencia , Proteínas ADAM/genética , Animales , Diferenciación Celular , Colitis/inducido químicamente , Colitis/genética , Colitis/patología , Colon/inmunología , Colon/patología , Matriz Extracelular/inmunología , Fibroblastos/patología , Fibrosis , Regulación de la Expresión Génica , Humanos , Inflamación , Interleucina-11/genética , Interleucina-11/inmunología , Mucosa Intestinal/patología , Masculino , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Dodecil Sulfato de Sodio/administración & dosificación , Transcripción Genética , Transcriptoma , Cicatrización de Heridas/genética , Cicatrización de Heridas/inmunología
7.
Cell ; 142(2): 320-32, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20655472

RESUMEN

A hallmark of Alzheimer's disease (AD) is the accumulation of plaques of Abeta 1-40 and 1-42 peptides, which result from the sequential cleavage of APP by the beta and gamma-secretases. The production of Abeta peptides is avoided by alternate cleavage of APP by the alpha and gamma-secretases. Here we show that production of beta-amyloid and plaques in a mouse model of AD are reduced by overexpressing the NAD-dependent deacetylase SIRT1 in brain, and are increased by knocking out SIRT1 in brain. SIRT1 directly activates the transcription of the gene encoding the alpha-secretase, ADAM10. SIRT1 deacetylates and coactivates the retinoic acid receptor beta, a known regulator of ADAM10 transcription. ADAM10 activation by SIRT1 also induces the Notch pathway, which is known to repair neuronal damage in the brain. Our findings indicate SIRT1 activation is a viable strategy to combat AD and perhaps other neurodegenerative diseases.


Asunto(s)
Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteínas de la Membrana/metabolismo , Sirtuina 1/metabolismo , Proteína ADAM10 , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurogénesis , Receptores Notch/metabolismo , Receptores de Ácido Retinoico/metabolismo , Tretinoina/metabolismo
8.
Circulation ; 147(9): 728-742, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36562301

RESUMEN

BACKGROUND: The metalloprotease ADAMTS-7 (a disintegrin and metalloproteinase with thrombospondin type 1 motif 7) is a novel locus associated with human coronary atherosclerosis. ADAMTS-7 deletion protects against atherosclerosis and vascular restenosis in rodents. METHODS: We designed 3 potential vaccines consisting of distinct B cell epitopic peptides derived from ADAMTS-7 and conjugated with the carrier protein KLH (keyhole limpet hemocyanin) as well as aluminum hydroxide as an adjuvant. Arterial ligation or wire injury was used to induce neointima in mice, whereas ApoE-/- and LDLR-/- (LDLR [low-density lipoprotein receptor]) mice fed a high-fat diet were applied to assess atherosclerosis. In addition, coronary stent implantation was performed on vaccine-immunized Bama miniature pigs, followed by optical coherence tomography to evaluate coronary intimal hyperplasia. RESULTS: A vaccine, ATS7vac, was screened out from 3 candidates to effectively inhibit intimal thickening in murine carotid artery ligation models after vaccination. As well, immunization with ATS7vac alleviated neointima formation in murine wire injury models and mitigated atherosclerotic lesions in both hyperlipidemic ApoE-/- and LDLR-/- mice without lowering lipid levels. Preclinically, ATS7vac markedly impeded intimal hyperplasia in swine stented coronary arteries, but without significant immune-related organ injuries. Mechanistically, ATS7vac vaccination produced specific antibodies against ADAMTS-7, which markedly repressed ADAMTS-7-mediated COMP (cartilage oligomeric matrix protein) and TSP-1 (thrombospondin-1) degradation and subsequently inhibited vascular smooth muscle cell migration but promoted re-endothelialization. CONCLUSIONS: ATS7vac is a novel atherosclerosis vaccine that also alleviates in-stent restenosis. The application of ATS7vac would be a complementary therapeutic avenue to the current lipid-lowering strategy for atherosclerotic disease.


Asunto(s)
Aterosclerosis , Neointima , Animales , Ratones , Proteínas ADAM/metabolismo , Aterosclerosis/patología , Modelos Animales de Enfermedad , Hiperplasia/metabolismo , Lípidos , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo , Porcinos , Trombospondinas/metabolismo , Vacunas de Subunidad/metabolismo , Proteína ADAMTS7
9.
Clin Immunol ; 262: 110168, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38458301

RESUMEN

Systemic lupus erythematosus (SLE) is a complex autoimmune disorder impacting various organs, notably prevalent in women of reproductive age. This review explores the involvement of a disintegrin and metalloproteinases (ADAMs) in SLE pathogenesis. Despite advancements in understanding SLE through genome and transcriptome studies, the role of ADAMs in post-translational regulations remains insufficiently explored. ADAMs, transmembrane proteins with diverse functions, impact cell adhesion, migration, and inflammation by shedding cell surface proteins, growth factors, and receptors. Notably, ADAM9 is implicated in Th17 cell differentiation, which is crucial in SLE pathology. ADAM10 and ADAM17 play pivotal roles in T-cell biology, influencing immune cell development and differentiation. Elevated soluble ADAM substrates in SLE patients serve as potential biomarkers correlating with disease activity. Targeting ADAMs or their substrates offers promising therapeutic avenues for SLE management and treatment enhancement.


Asunto(s)
Desintegrinas , Lupus Eritematoso Sistémico , Humanos , Femenino , Desintegrinas/metabolismo , Proteína ADAM10/metabolismo , Inflamación , Diferenciación Celular , Proteínas de la Membrana , Proteínas ADAM
10.
Clin Immunol ; 263: 110228, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38663494

RESUMEN

Asthma is a heterogeneous disease characterized by chronic airway inflammation, reversible airflow limitation, and airway remodeling. Eosinophil peroxidase (EPX) is the most abundant secondary granule protein unique to activated eosinophils. In this study, we aimed to illustrate the effect of EPX on the epithelial-mesenchymal transition (EMT) in BEAS-2B cells. Our research found that both EPX and ADAM33 were negatively correlated with FEV1/FVC and FEV1%pred, and positively correlated with IL-5 levels. Asthma patients had relatively higher levels of ADAM33 and EPX compared to the healthy control group. The expression of TSLP, TGF-ß1 and ADAM33 in the EPX intervention group was significantly higher. Moreover, EPX could promote the proliferation, migration and EMT of BEAS-2B cells, and the effect of EPX on various factors was significantly improved by the PI3K inhibitor LY294002. The findings from this study could potentially offer a novel therapeutic target for addressing airway remodeling in bronchial asthma, particularly focusing on EMT.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Asma , Bronquios , Peroxidasa del Eosinófilo , Células Epiteliales , Transición Epitelial-Mesenquimal , Factor de Crecimiento Transformador beta1 , Humanos , Asma/metabolismo , Asma/patología , Asma/fisiopatología , Asma/inmunología , Masculino , Femenino , Células Epiteliales/metabolismo , Peroxidasa del Eosinófilo/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Persona de Mediana Edad , Adulto , Bronquios/patología , Interleucina-5/metabolismo , Cromonas/farmacología , Citocinas/metabolismo , Línea Celular , Linfopoyetina del Estroma Tímico , Proliferación Celular , Movimiento Celular , Morfolinas/farmacología , Proteínas ADAM
11.
EMBO J ; 39(20): e105693, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32954517

RESUMEN

To understand how cells communicate in the nervous system, it is essential to define their secretome, which is challenging for primary cells because of large cell numbers being required. Here, we miniaturized secretome analysis by developing the "high-performance secretome protein enrichment with click sugars" (hiSPECS) method. To demonstrate its broad utility, hiSPECS was used to identify the secretory response of brain slices upon LPS-induced neuroinflammation and to establish the cell type-resolved mouse brain secretome resource using primary astrocytes, microglia, neurons, and oligodendrocytes. This resource allowed mapping the cellular origin of CSF proteins and revealed that an unexpectedly high number of secreted proteins in vitro and in vivo are proteolytically cleaved membrane protein ectodomains. Two examples are neuronally secreted ADAM22 and CD200, which we identified as substrates of the Alzheimer-linked protease BACE1. hiSPECS and the brain secretome resource can be widely exploited to systematically study protein secretion and brain function and to identify cell type-specific biomarkers for CNS diseases.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Oligodendroglía/metabolismo , Proteómica/métodos , Programas Informáticos , Proteínas ADAM/líquido cefalorraquídeo , Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/líquido cefalorraquídeo , Animales , Antígenos CD/líquido cefalorraquídeo , Antígenos CD/metabolismo , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/líquido cefalorraquídeo , Encéfalo/citología , Células Cultivadas , Proteínas del Líquido Cefalorraquídeo , Cromatografía Liquida , Ontología de Genes , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/líquido cefalorraquídeo , Proteínas del Tejido Nervioso/metabolismo , Análisis de Componente Principal , Proteoma/metabolismo , Espectrometría de Masas en Tándem
12.
Am J Physiol Heart Circ Physiol ; 327(2): H409-H416, 2024 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-38607341

RESUMEN

Cardiac hypertrophy is a common feature in several cardiomyopathies. We previously reported that loss of ADAM15 (disintegrin and metalloproteinase 15) worsened cardiac hypertrophy and dilated cardiomyopathy following cardiac pressure overload. Here, we investigated the impact of ADAM15 loss in female mice following cardiac pressure overload induced by transverse aortic constriction (TAC). Female Adam15-/- mice developed the same degree of cardiac hypertrophy, dilation, and dysfunction as the parallel female wild-type (WT) mice at 6 wk post-TAC. To determine if this is due to the protective effects of estrogen, which could mask the negative impact of Adam15 loss, WT and Adam15-/- mice underwent ovariectomy (OVx) 2 wk before TAC. Cardiac structure and function analyses were performed at 6 wk post-TAC. OVx similarly impacted females of both genotypes post-TAC. Calcineurin (Cn) activity was increased post-OVx-TAC, and more in Adam15-/- mice; however, this increase was not reflected in the total-to-phospho-NFAT levels. Integrin-α7 expression, which was upstream of Cn activation in male Adam15-/- -TAC mice, remained unchanged in female mice. However, activation of the mitogen-activated protein kinases (ERK, JNK, P38) was greater in Adam15-/--OVx-TAC than in WT-OVx-TAC mice. In addition, ADAM15 protein levels were significantly increased post-TAC in male but not in female WT mice. Myocardial fibrosis was comparable in non-OVx WT-TAC and Adam15-/- -TAC mice. OVx increased the perivascular fibrosis more in Adam15-/- compared with WT mice post-TAC. Our data demonstrate that loss of ovarian hormones did not fully replicate the male phenotype in the female Adam15-/- mice post-TAC. As ADAM15 levels were increased in males but not in females post-TAC, it is plausible that ADAM15 does not play a prominent role in post-TAC events in female mice. Our findings highlight the significance of factors other than sex hormones in mediating cardiomyopathies in females, which require a more thorough understanding.NEW & NOTEWORTHY Loss of ADAM15 in female mice, unlike the male mice, does not worsen the cardiomyopathy following cardiac pressure overload. Ovariectomy does not worsen the post-TAC cardiomyopathy in female Adam15-/- mice compared with female WT mice. Lack of deleterious impact of Adam15 deficiency in female mice is not because of the protective effects of ovarian hormones but could be due to a less prominent role of ADAM15 in cardiac response to post-TAC remodeling in female mice.


Asunto(s)
Proteínas ADAM , Proteínas de la Membrana , Ratones Noqueados , Ovariectomía , Animales , Femenino , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteínas ADAM/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Masculino , Ratones Endogámicos C57BL , Calcineurina/metabolismo , Calcineurina/genética , Modelos Animales de Enfermedad , Ratones , Remodelación Ventricular , Factores de Transcripción NFATC/metabolismo , Factores de Transcripción NFATC/genética , Miocardio/metabolismo , Miocardio/patología , Fibrosis , Cardiomiopatías/fisiopatología , Cardiomiopatías/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/etiología , Cardiomiopatías/patología , Factores Sexuales , Transducción de Señal , Función Ventricular Izquierda , Hipertrofia Ventricular Izquierda/fisiopatología , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/etiología
13.
Biol Reprod ; 110(4): 750-760, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38217862

RESUMEN

Sperm proteins undergo post-translational modifications during sperm transit through the epididymis to acquire fertilizing ability. We previously reported that the genomic region coding Pate family genes is key to the proteolytic processing of the sperm membrane protein ADAM3 and male fertility. This region contains nine Pate family genes (Pate5-13), and two protein-coding genes (Gm27235 and Gm5916), with a domain structure similar to Pate family genes. Therefore, in this study, we aimed to identify key factors by narrowing the genomic region. We generated three knockout (KO) mouse lines using CRISPR/Cas9: single KO mice of Pate10 expressed in the caput epididymis; deletion KO mice of six caput epididymis-enriched genes (Pate5-7, 13, Gm27235, and Gm5916) (Pate7-Gm5916 KO); and deletion KO mice of four genes expressed in the placenta and epididymis (Pate8, 9, 11, and 12) (Pate8-12 KO). We observed that the fertility of only Pate7-Gm5916 KO males was reduced, whereas the rest remained unaffected. Furthermore, when the caput epididymis-enriched genes, Pate8 and Pate10 remained in Pate7-Gm5916 KO mice were independently deleted, both KO males displayed more severe subfertility due to a decrease in mature ADAM3 and a defect in sperm migration to the oviduct. Thus, our data showed that multiple caput epididymis-enriched genes within the region coding Pate5-13 cooperatively function to ensure male fertility in mice.


Asunto(s)
Proteínas ADAM , Espermatozoides , Animales , Femenino , Masculino , Ratones , Embarazo , Epidídimo/metabolismo , Fertilidad/genética , Genómica , Ratones Noqueados , Semen , Espermatozoides/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo
14.
Immunity ; 42(4): 756-66, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25902485

RESUMEN

Staphylococcus aureus skin colonization is universal in atopic dermatitis and common in cancer patients treated with epidermal growth factor receptor inhibitors. However, the causal relationship of dysbiosis and eczema has yet to be clarified. Herein, we demonstrate that Adam17(fl/fl)Sox9-(Cre) mice, generated to model ADAM17-deficiency in human, developed eczematous dermatitis with naturally occurring dysbiosis, similar to that observed in atopic dermatitis. Corynebacterium mastitidis, S. aureus, and Corynebacterium bovis sequentially emerged during the onset of eczematous dermatitis, and antibiotics specific for these bacterial species almost completely reversed dysbiosis and eliminated skin inflammation. Whereas S. aureus prominently drove eczema formation, C. bovis induced robust T helper 2 cell responses. Langerhans cells were required for eliciting immune responses against S. aureus inoculation. These results characterize differential contributions of dysbiotic flora during eczema formation, and highlight the microbiota-host immunity axis as a possible target for future therapeutics in eczematous dermatitis.


Asunto(s)
Dermatitis Atópica/inmunología , Disbiosis/inmunología , Eccema/inmunología , Células de Langerhans/inmunología , Piel/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Proteínas ADAM/deficiencia , Proteínas ADAM/genética , Proteínas ADAM/inmunología , Proteína ADAM17 , Animales , Antibacterianos/farmacología , Corynebacterium/inmunología , Dermatitis Atópica/tratamiento farmacológico , Dermatitis Atópica/genética , Dermatitis Atópica/microbiología , Disbiosis/tratamiento farmacológico , Disbiosis/genética , Disbiosis/microbiología , Eccema/tratamiento farmacológico , Eccema/genética , Eccema/microbiología , Receptores ErbB/genética , Receptores ErbB/inmunología , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/inmunología , Inflamación/microbiología , Integrasas/genética , Integrasas/inmunología , Células de Langerhans/efectos de los fármacos , Células de Langerhans/microbiología , Células de Langerhans/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/inmunología , Transducción de Señal , Piel/efectos de los fármacos , Piel/microbiología , Piel/patología , Staphylococcus aureus/inmunología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/microbiología , Linfocitos T Colaboradores-Inductores/patología
15.
J Med Primatol ; 53(1): e12686, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37990472

RESUMEN

We performed whole-exome sequencing using a human exome capture kit to analyze the potential genetic factors related to patent ductus arteriosus in Japanese macaques. Compared with the reference sequences of other primates, we identified potential missense variants in five genes: ADAM15, AZGP1, CSPG4, TNFRSF13B, and EPOR.


Asunto(s)
Conducto Arterioso Permeable , Humanos , Animales , Conducto Arterioso Permeable/genética , Conducto Arterioso Permeable/veterinaria , Macaca fuscata , Secuenciación del Exoma , Proteínas de la Membrana/genética , Proteínas ADAM/genética
16.
Scand J Gastroenterol ; 59(3): 304-315, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37978827

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is the second leading cause of cancer-related death. Immunotherapy is one of the new options for cancer treatment. This study aimed to develop an immune-related signature associated with CRC. METHODS: We performed differential analysis to screen out the differentially expressed genes (DEGs) of The Cancer Genome Atlas-Colorectal Cancer (TCGA-CRC) datasets. Weighted gene co-expression network analysis (WGCNA) was performed to obtain the key module genes associated with differential immune cells. The candidate genes were obtained through overlapping key DEGs and key module genes. The univariate and multivariate Cox regression analyses were adopted to build a CRC prognostic signature. We further conducted immune feature estimation and chemotherapy analysis between two risk subgroups. Finally, we verified the expression of immune-related prognostic genes at the transcriptional level. RESULTS: A total of 61 candidate genes were obtained by overlapping key DEGs and key module genes associated with differential immune cells. Then, an immune-related prognostic signature was built based on the three prognostic genes (HAMP, ADAM8, and CD1B). The independent prognostic analysis suggested that age, stage, and RiskScore could be used as independent prognostic factors. Further, we found significantly higher expression of three prognostic genes in the CRC group compared with the normal group. Finally, real-time polymerase chain reaction verified the expression of three genes in patients with CRC. CONCLUSION: The prognostic signature comprising HAMP, ADAM8, and CD1B based on immune cells was established, providing a theoretical basis and reference value for the research of CRC.


Asunto(s)
Neoplasias Colorrectales , Microambiente Tumoral , Humanos , Pronóstico , Microambiente Tumoral/genética , Expresión Génica , Perfilación de la Expresión Génica , Neoplasias Colorrectales/genética , Proteínas de la Membrana , Proteínas ADAM
17.
Biotechnol Appl Biochem ; 71(1): 45-60, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37881150

RESUMEN

Angiogenesis is associated with tumor progression, prognosis, and treatment effect. However, the angiogenesis' underlying mechanisms in the tumor microenvironment (TME) still remain unclear. Understanding the dynamic interactions between angiogenesis and TME in colon adenocarcinoma (COAD) is necessary. We downloaded the transcriptome data and corresponding clinical data of colon cancer patients from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases, respectively. We identified two distinct angiogenesis-related molecular subtypes (subtype A and subtype B) and assessed the clinical features, prognosis, and infiltrating immune cells of patients in the two subtypes. According to the prognostic differential genes, we defined two different gene clusters to further explore the correlation between angiogenesis and tumor heterogeneity. Then, we construct the prognostic risk scoring model angiogenesis-related gene (ARG-score) including seven genes (ARMCX2, latent transforming growth factor ß binding protein 1, ADAM8, FABP4, CCL11, CXCL11, ITLN1) using Lasso-multivariate cox method. We analyzed the correlation between ARG-score and prognosis, clinicopathological features, TME, molecular feature, cancer stem cells (CSCs), and microsatellite instability (MSI) status. To assess the application value of ARG-score in clinical treatment, immunophenotype score was used to predict patients' immunotherapy response in colon cancer. We found the mutations of ARGs in TCGA-COAD dataset from genetic levels and discussed their expression patterns based on TCGA and GEO datasets. We observed important differences in clinicopathological features, prognosis, immune feature, molecular feature between the two molecular subgroups. Then, we established an ARG-score for predicting OS and validated its predictive capability. A high ARG-score characterized by higher transcription level of ARGs, suggested lower MSI-high (MSI-H), lower immune score, and worse clinical stage and survival outcome. Additionally, the ARG-score was remarkably related to the CSCs index and immunotherapy sensitivity. We found two new molecular subtypes and two gene clusters based on ARGs and established an ARG-score. Multilayered analysis revealed that ARGs were remarkably correlated to the heterogeneity of colon cancer patients and explained the process of tumorigenesis and progression better. The ARG-score can help us better assess patients' survival outcomes and provide guidance for individualized treatment.


Asunto(s)
Adenocarcinoma , Neoplasias del Colon , Humanos , Neoplasias del Colon/genética , Microambiente Tumoral/genética , Angiogénesis , Pronóstico , Proteínas de la Membrana , Proteínas ADAM
18.
Cell Mol Biol Lett ; 29(1): 75, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755530

RESUMEN

BACKGROUND: Mechanical spinal cord injury (SCI) is a deteriorative neurological disorder, causing secondary neuroinflammation and neuropathy. ADAM8 is thought to be an extracellular metalloproteinase, which regulates proteolysis and cell adherence, but whether its intracellular region is involved in regulating neuroinflammation in microglia after SCI is unclear. METHODS: Using animal tissue RNA-Seq and clinical blood sample examinations, we found that a specific up-regulation of ADAM8 in microglia was associated with inflammation after SCI. In vitro, microglia stimulated by HMGB1, the tail region of ADAM8, promoted microglial inflammation, migration and proliferation by directly interacting with ERKs and Fra-1 to promote activation, then further activated Map3k4/JNKs/p38. Using SCI mice, we used BK-1361, a specific inhibitor of ADAM8, to treat these mice. RESULTS: The results showed that administration of BK-1361 attenuated the level of neuroinflammation and reduced microglial activation and recruitment by inhibiting the ADAM8/Fra-1 axis. Furthermore, treatment with BK-1361 alleviated glial scar formation, and also preserved myelin and axonal structures. The locomotor recovery of SCI mice treated with BK-1361 was therefore better than those without treatment. CONCLUSIONS: Taken together, the results showed that ADAM8 was a critical molecule, which positively regulated neuroinflammatory development and secondary pathogenesis by promoting microglial activation and migration. Mechanically, ADAM8 formed a complex with ERK and Fra-1 to further activate the Map3k4/JNK/p38 axis in microglia. Inhibition of ADAM8 by treatment with BK-1361 decreased the levels of neuroinflammation, glial formation, and neurohistological loss, leading to favorable improvement in locomotor functional recovery in SCI mice.


Asunto(s)
Proteínas ADAM , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana , Microglía , Enfermedades Neuroinflamatorias , Traumatismos de la Médula Espinal , Animales , Ratones , Proteínas ADAM/metabolismo , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/genética , Antígenos CD , Movimiento Celular/efectos de los fármacos , Inflamación/patología , Inflamación/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/efectos de los fármacos , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/tratamiento farmacológico
19.
Skin Res Technol ; 30(7): e13630, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38988131

RESUMEN

OBJECTIVE: To investigate the role of NEAT1 targeted regulation of miR-125/ADAM9 mediated NF-κB pathway in inflammatory response in rosacea. METHOD: HaCaT cell rosacea phenotype was induced by LL37. The connection targeted by NEAT1 and miR-125a-5p was confirmed by Double-Luciferase report analysis. qPCR was employed to assess the levels of expression for NEAT1, miR-125a-5p, and ADAM9 genes. The levels of expression for ADAM9/TLR2/NF-κB P65 pathway proteins in each batch of cells were determined by Western blotting. The levels of expression for inflammatory factors, including TNF-α, IL-1ß, IL-6, and IL-18, were measured through ELISA experimentation. RESULTS: LL37 could successfully induce HaCaT cells to exhibit rosacea phenotype. The luciferase report experiment confirmed that NEAT1 could target and bind miR-125a-5p and inhibit its expression. ADAM9 exhibited increased expression in LL37-induced HaCaT cells, showing a positive association with NEAT1 expression and inverse relationship with miR-125a-5p activation. LL37 treatment promoted the expression of ADAM9/TLR2/NF-κB P65 pathway proteins. Silencing ADAM9 can inhibit the inflammatory signaling pathway and reduce the level of TNF-α, IL-1ß, IL-6, and IL-18 in HaCaT cells. CONCLUSION: NEAT1 can suppress the production of miR-125a-5p and activate the TLR2/NF-κB inflammatory pathway mediated by ADAM9, thereby promoting the inflammatory response in rosacea.


Asunto(s)
Proteínas ADAM , Proteínas de la Membrana , MicroARNs , FN-kappa B , ARN Largo no Codificante , Rosácea , Humanos , MicroARNs/metabolismo , MicroARNs/genética , Rosácea/metabolismo , Rosácea/genética , Proteínas ADAM/metabolismo , Proteínas ADAM/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , FN-kappa B/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Transducción de Señal , Células HaCaT , Catelicidinas , Péptidos Catiónicos Antimicrobianos/metabolismo , Péptidos Catiónicos Antimicrobianos/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 2/genética
20.
PLoS Genet ; 17(5): e1009506, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33956822

RESUMEN

Identifying the molecular underpinnings of the neural specializations that underlie human cognitive and behavioral traits has long been of considerable interest. Much research on human-specific changes in gene expression and epigenetic marks has focused on the prefrontal cortex, a brain structure distinguished by its role in executive functions. The cerebellum shows expansion in great apes and is gaining increasing attention for its role in motor skills and cognitive processing, including language. However, relatively few molecular studies of the cerebellum in a comparative evolutionary context have been conducted. Here, we identify human-specific methylation in the lateral cerebellum relative to the dorsolateral prefrontal cortex, in a comparative study with chimpanzees (Pan troglodytes) and rhesus macaques (Macaca mulatta). Specifically, we profiled genome-wide methylation levels in the three species for each of the two brain structures and identified human-specific differentially methylated genomic regions unique to each structure. We further identified which differentially methylated regions (DMRs) overlap likely regulatory elements and determined whether associated genes show corresponding species differences in gene expression. We found greater human-specific methylation in the cerebellum than the dorsolateral prefrontal cortex, with differentially methylated regions overlapping genes involved in several conditions or processes relevant to human neurobiology, including synaptic plasticity, lipid metabolism, neuroinflammation and neurodegeneration, and neurodevelopment, including developmental disorders. Moreover, our results show some overlap with those of previous studies focused on the neocortex, indicating that such results may be common to multiple brain structures. These findings further our understanding of the cerebellum in human brain evolution.


Asunto(s)
Cerebelo/metabolismo , Metilación de ADN , Epigénesis Genética , Proteínas ADAM , Animales , Autoantígenos , Proteínas Portadoras , Chad , Islas de CpG , Femenino , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Macaca mulatta/genética , Masculino , Proteínas de Microfilamentos , Proteínas del Tejido Nervioso , Pan troglodytes/genética , Fosfoinositido Fosfolipasa C , Proteínas Serina-Treonina Quinasas , Proteínas , Proteínas Asociadas a SAP90-PSD95 , Especificidad de la Especie , Sitio de Iniciación de la Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA