Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Cell Physiol ; 323(3): C651-C665, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35785985

RESUMEN

Proteoglycans are composite molecules comprising a protein backbone, i.e., the core protein, with covalently attached glycosaminoglycan chains of distinct chemical types. Most proteoglycans are secreted or attached to the cell membrane. Their specialized structures, binding properties, and biophysical attributes underlie diverse biological roles, which include modulation of tissue mechanics, cell adhesion, and the sequestration and regulated release of morphogens, growth factors, and cytokines. As an irreversible post-translational modification, proteolysis has a profound impact on proteoglycan function, abundance, and localization. Proteolysis is required for molecular maturation of some proteoglycans, clearance of extracellular matrix proteoglycans during tissue remodeling, generation of bioactive fragments from proteoglycans, and ectodomain shedding of cell-surface proteoglycans. Genetic evidence shows that proteoglycan core protein proteolysis is essential for diverse morphogenetic events during embryonic development. In contrast, dysregulated proteoglycan proteolysis contributes to osteoarthritis, cardiovascular disorders, cancer, and inflammation. Proteolytic fragments of perlecan, versican, aggrecan, brevican, collagen XVIII, and other proteoglycans are associated with independent biological activities as so-called matrikines. Yet, proteoglycan proteolysis has been investigated to only a limited extent to date. Here, we review the actions of proteases on proteoglycans and illustrate their functional impact with several examples. We discuss the applications and limitations of strategies used to define cleavage sites in proteoglycans and explain how proteoglycanome-wide proteolytic mapping, which is desirable to fully understand the impact of proteolysis on proteoglycans, can be facilitated by integrating classical proteoglycan isolation methods with mass spectrometry-based proteomics.


Asunto(s)
Matriz Extracelular , Versicanos , Agrecanos/metabolismo , Matriz Extracelular/metabolismo , Procesamiento Proteico-Postraduccional , Proteolisis , Versicanos/metabolismo
2.
J Hepatol ; 74(4): 893-906, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33197513

RESUMEN

BACKGROUND & AIMS: The tumour microenvironment shapes tumour growth through cellular communications that include both direct interactions and secreted factors. The aim of this study was to characterize the impact of the secreted glycoprotein ADAMTSL5, whose role in cancer has not been previously investigated, on hepatocellular carcinoma (HCC). METHODS: ADAMTSL5 methylation status was evaluated through bisulfite sequencing, and publicly available data analysis. ADAMTSL5 RNA and protein expression were assessed in mouse models and HCC patient samples and compared to data from published datasets. Functional studies, including association of ADAMTSL5 depletion with responsiveness to clinically relevant drugs, were performed in cellular and in vivo models. Molecular alterations associated with ADAMTSL5 targeting were determined using proteomics, biochemistry, and reverse-transcription quantitative PCR. RESULTS: Methylome analysis revealed hypermethylated gene body CpG islands at the ADAMTSL5 locus in both mouse and human HCC, correlating with higher ADAMTSL5 expression. ADAMTSL5 targeting interfered with tumorigenic properties of HCC cells in vitro and in vivo, whereas ADAMTSL5 overexpression conferred tumorigenicity to pre-tumoural hepatocytes sensitized to transformation by a modest level of MET receptor expression. Mechanistically, ADAMTSL5 abrogation led to a reduction of several oncogenic inputs relevant to HCC, including reduced expression and/or phosphorylation levels of receptor tyrosine kinases MET, EGFR, PDGFRß, IGF1Rß, or FGFR4. This phenotype was associated with significantly increased sensitivity of HCC cells to clinically relevant drugs, namely sorafenib, lenvatinib, and regorafenib. Moreover, ADAMTSL5 depletion drastically increased expression of AXL, accompanied by a sensitization to bemcentinib. CONCLUSIONS: Our results point to a role for ADAMTSL5 in maintaining the function of key oncogenic signalling pathways, suggesting that it may act as a master regulator of tumorigenicity and drug resistance in HCC. LAY SUMMARY: The environment of cancer cells has profound effects on establishment, progression, and response of a tumour to treatment. Herein, we show that ADAMTSL5, a protein secreted by liver cancer cells and overlooked in cancer so far, is increased in this tumour type, is necessary for tumour formation and supports drug resistance. Adamtsl5 removal conferred sensitivity of liver cancer cells to drugs used in current treatment. This suggests ADAMTSL5 as a potential marker in liver cancer as well as a possible drug target.


Asunto(s)
Proteínas ADAMTS , Proteína ADAMTS5 , Carcinogénesis , Carcinoma Hepatocelular , Resistencia a Antineoplásicos/fisiología , Neoplasias Hepáticas , Transducción de Señal , Proteínas ADAMTS/genética , Proteínas ADAMTS/metabolismo , Proteína ADAMTS5/genética , Proteína ADAMTS5/metabolismo , Animales , Antineoplásicos Inmunológicos/farmacología , Benzocicloheptenos/farmacología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Epigenómica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Compuestos de Fenilurea/farmacología , Quinolinas/farmacología , Sorafenib/farmacología , Activación Transcripcional , Triazoles/farmacología , Microambiente Tumoral/fisiología
3.
4.
Nucleic Acids Res ; 43(11): 5394-408, 2015 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-25940622

RESUMEN

Two decades after the discovery that heterozygous mutations within and around SOX9 cause campomelic dysplasia, a generalized skeleton malformation syndrome, it is well established that SOX9 is a master transcription factor in chondrocytes. In contrast, the mechanisms whereby translocations in the --350/-50-kb region 5' of SOX9 cause severe disease and whereby SOX9 expression is specified in chondrocytes remain scarcely known. We here screen this upstream region and uncover multiple enhancers that activate Sox9-promoter transgenes in the SOX9 expression domain. Three of them are primarily active in chondrocytes. E250 (located at -250 kb) confines its activity to condensed prechondrocytes, E195 mainly targets proliferating chondrocytes, and E84 is potent in all differentiated chondrocytes. E84 and E195 synergize with E70, previously shown to be active in most Sox9-expressing somatic tissues, including cartilage. While SOX9 protein powerfully activates E70, it does not control E250. It requires its SOX5/SOX6 chondrogenic partners to robustly activate E195 and additional factors to activate E84. Altogether, these results indicate that SOX9 expression in chondrocytes relies on widely spread transcriptional modules whose synergistic and overlapping activities are driven by SOX9, SOX5/SOX6 and other factors. They help elucidate mechanisms underlying campomelic dysplasia and will likely help uncover other disease mechanisms.


Asunto(s)
Condrocitos/metabolismo , Elementos de Facilitación Genéticos , Factor de Transcripción SOX9/genética , Activación Transcripcional , Animales , Células COS , Displasia Campomélica/genética , Linaje de la Célula , Células Cultivadas , Chlorocebus aethiops , Condrocitos/citología , Aberraciones Cromosómicas , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Factores de Transcripción SOXD
5.
Nucleic Acids Res ; 41(8): 4459-69, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23449223

RESUMEN

SOX9 encodes a transcription factor that presides over the specification and differentiation of numerous progenitor and differentiated cell types, and although SOX9 haploinsufficiency and overexpression cause severe diseases in humans, including campomelic dysplasia, sex reversal and cancer, the mechanisms underlying SOX9 transcription remain largely unsolved. We identify here an evolutionarily conserved enhancer located 70-kb upstream of mouse Sox9 and call it SOM because it specifically activates a Sox9 promoter reporter in most Sox9-expressing somatic tissues in transgenic mice. Moreover, SOM-null fetuses and pups reduce Sox9 expression by 18-37% in the pancreas, lung, kidney, salivary gland, gut and liver. Weanlings exhibit half-size pancreatic islets and underproduce insulin and glucagon, and adults slowly recover from acute pancreatitis due to a 2-fold impairment in Sox9 upregulation. Molecular and genetic experiments reveal that Sox9 protein dimers bind to multiple recognition sites in the SOM sequence and are thereby both necessary and sufficient for enhancer activity. These findings thus uncover that Sox9 directly enhances its functions in somatic tissue development and adult regeneration through SOM-mediated positive auto-regulation. They provide thereby novel insights on molecular mechanisms controlling developmental and disease processes and suggest new strategies to improve disease treatments.


Asunto(s)
Elementos de Facilitación Genéticos , Regeneración , Factor de Transcripción SOX9/genética , Animales , Línea Celular , Embrión de Mamíferos/metabolismo , Homeostasis , Ratones , Ratones Transgénicos , Páncreas/crecimiento & desarrollo , Páncreas/fisiología , Pancreatitis/patología , Ratas , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/fisiología
6.
Matrix Biol ; 131: 1-16, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38750698

RESUMEN

Extracellular matrix remodeling mechanisms are understudied in cardiac development and congenital heart defects. We show that matrix-degrading metalloproteases ADAMTS1 and ADAMTS5, are extensively co-expressed during mouse cardiac development. The mouse mutants of each gene have mild cardiac anomalies, however, their combined genetic inactivation to elicit cooperative roles is precluded by tight gene linkage. Therefore, we coupled Adamts1 inactivation with pharmacologic ADAMTS5 blockade to uncover stage-specific cooperative roles and investigated their potential substrates in mouse cardiac development. ADAMTS5 blockade was achieved in Adamts1 null mouse embryos using an activity-blocking monoclonal antibody during distinct developmental windows spanning myocardial compaction or cardiac septation and outflow tract rotation. Synchrotron imaging, RNA in situ hybridization, immunofluorescence microscopy and electron microscopy were used to determine the impact on cardiac development and compared to Gpc6 and ADAMTS-cleavage resistant versican mutants. Mass spectrometry-based N-terminomics was used to seek relevant substrates. Combined inactivation of ADAMTS1 and ADAMTS5 prior to 12.5 days of gestation led to dramatic accumulation of versican-rich cardiac jelly and inhibited formation of compact and trabecular myocardium, which was also observed in mice with ADAMTS cleavage-resistant versican. Combined inactivation after 12.5 days impaired outflow tract development and ventricular septal closure, generating a tetralogy of Fallot-like defect. N-terminomics of combined ADAMTS knockout and control hearts identified a cleaved glypican-6 peptide only in the controls. ADAMTS1 and ADAMTS5 expression in cells was associated with specific glypican-6 cleavages. Paradoxically, combined ADAMTS1 and ADAMTS5 inactivation reduced cardiac glypican-6 and outflow tract Gpc6 transcription. Notably, Gpc6-/- hearts demonstrated similar rotational defects as combined ADAMTS inactivated hearts and both had reduced hedgehog signaling. Thus, versican proteolysis in cardiac jelly at the canonical Glu441-Ala442 site is cooperatively mediated by ADAMTS1 and ADAMTS5 and required for proper ventricular cardiomyogenesis, whereas, reduced glypican-6 after combined ADAMTS inactivation impairs hedgehog signaling, leading to outflow tract malrotation.


Asunto(s)
Proteína ADAMTS1 , Proteína ADAMTS5 , Glipicanos , Corazón , Proteolisis , Versicanos , Animales , Ratones , Versicanos/metabolismo , Versicanos/genética , Proteína ADAMTS5/metabolismo , Proteína ADAMTS5/genética , Proteína ADAMTS1/metabolismo , Proteína ADAMTS1/genética , Glipicanos/metabolismo , Glipicanos/genética , Corazón/crecimiento & desarrollo , Ratones Noqueados , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/patología
7.
Dev Dyn ; 241(2): 376-89, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22275227

RESUMEN

BACKGROUND: The function of Notch signaling in murine neural crest-derived cell lineages in vivo was examined. RESULTS: Conditional gain (Wnt1Cre;Rosa(Notch)) or loss (Wnt1Cre;RBP-J(f/f)) of Notch signaling in neural crest cells (NCCs) in vivo results in craniofacial, cardiac, and trunk abnormalities. Severe craniofacial malformations are apparent in Wnt1Cre;Rosa(Notch) embryos, while less severe skull abnormalities are evident in Wnt1Cre;RBP-J(f/f) mice. Deficient cardiac neural crest migration, resulting in cardiac outflow tract malformations, occurs with increased or decreased Notch signaling in NCCs. Smooth muscle cell differentiation also is impaired in pharyngeal NCC derivatives in both Wnt1Cre;Rosa(Notch) and Wnt1Cre;RBP-J(f/f) embryos. Neurogenesis is absent and gliogenesis is increased in the dorsal root ganglia of Wnt1Cre;Rosa(Notch) embryos, while neurogenesis is increased and gliogenesis is decreased in Wnt1Cre;RBP-J(f/f) embryos. CONCLUSIONS: Together, these studies demonstrate essential cell-autonomous roles for appropriate levels of Notch signaling during NCC migration, proliferation, and differentiation with critical implications in craniofacial, cardiac, and neurogenic development and disease.


Asunto(s)
Diferenciación Celular , Cresta Neural/citología , Cresta Neural/embriología , Receptores Notch/metabolismo , Animales , Región Branquial/irrigación sanguínea , Región Branquial/embriología , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Cardiopatías Congénitas/genética , Ratones , Ratones Mutantes , Miocitos del Músculo Liso/fisiología , Receptores Notch/genética , Transducción de Señal , Proteína Wnt1/metabolismo
8.
Dev Dyn ; 241(3): 493-504, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22275127

RESUMEN

BACKGROUND: During vertebrate lens development, the lens placode in the embryonic ectoderm invaginates into a lens vesicle, which then separates from the surface epithelium, followed by two waves of fiber cell differentiation. In the mouse, multiple labs have shown that Jag1-Notch signaling is critically required during the second wave of lens fiber cell formation. However, Notch signaling appears to play no obvious role during lens induction or morphogenesis, although multiple pathway genes are expressed at these earlier stages. RESULTS: Here, we explored functions for Notch signaling specifically during early lens development, by using the early-acting AP2α-Cre driver to delete Jag1 or Rbpj. We found that Jag1 and Rbpj are not required during lens induction, but are necessary for proper lens vesicle separation from the surface ectoderm. CONCLUSIONS: We conclude that precise levels of Notch signaling are essential during lens vesicle morphogenesis. In addition, AP2α-Cre-mediated deletion of Rbpj resulted in embryos with cardiac outflow tract and liver deformities, and perinatal lethality.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Cristalino/embriología , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Complejo 2 de Proteína Adaptadora/genética , Animales , Proteínas de Unión al Calcio/genética , Eliminación de Gen , Cardiopatías Congénitas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Integrasas/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Proteína Jagged-1 , Cristalino/metabolismo , Hígado/anomalías , Hígado/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Proteínas Serrate-Jagged , Transducción de Señal
9.
Pulm Circ ; 13(1): e12200, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36824691

RESUMEN

Expansion of extracellular matrix occurs in all stages of pulmonary angiopathy associated with pulmonary arterial hypertension (PAH). In systemic arteries, dysregulation and accumulation of the large chondroitin-sulfate proteoglycan aggrecan is associated with swelling and disruption of vessel wall homeostasis. Whether aggrecan is present in pulmonary arteries, and its potential roles in PAH, has not been thoroughly investigated. Here, lung tissue from 11 patients with idiopathic PAH was imaged using synchrotron radiation phase-contrast microcomputed tomography (TOMCAT beamline, Swiss Light Source). Immunohistochemistry for aggrecan core protein in subsequently sectioned lung tissue demonstrated accumulation in PAH compared with failed donor lung controls. RNAscope in situ hybridization indicated ACAN expression in vascular endothelium and smooth muscle cells. Based on qualitative histological analysis, aggrecan localizes to cellular, rather than fibrotic or collagenous, lesions. Interestingly, ADAMTS15, a potential aggrecanase, was upregulated in pulmonary arteries in PAH. Aligning traditional histological analysis with three-dimensional renderings of pulmonary arteries from synchrotron imaging identified aggrecan in lumen-reducing lesions containing loose, cell-rich connective tissue, at sites of intrapulmonary bronchopulmonary shunting, and at sites of presumed elevated pulmonary blood pressure. Our findings suggest that ACAN expression may be an early response to injury in pulmonary angiopathy and supports recent work showing that dysregulation of aggrecan turnover is a hallmark of arterial adaptations to altered hemodynamics. Whether cause or effect, aggrecan and aggrecanase regulation in PAH are potential therapeutic targets.

10.
Adv Exp Med Biol ; 727: 114-30, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22399343

RESUMEN

Notch signaling is an important regulator of skeletogenesis at multiple developmental stages. The Notch signaling pathway is involved in the promotion of somite segmentation, patterning and differentiation into sclerotome pre-chondrogenic cells to allow for appropriate axial skeleton development. In addition, studies performed in vitro and in vivo demonstrate that Notch signaling suppresses chondrogenic and osteoblastic differentiation and negatively regulates osteoclast formation and proliferation. Through the use of in vitro and in vivo approaches, Notch signaling has been shown to regulate somitogenesis, chondrogenesis, osteoblastogenesis and osteoclastogenesis that ultimately affect skeletogenesis. Dysregulation of Notch signaling results in congenital skeletal malformations that could reveal therapeutic potential.


Asunto(s)
Enfermedades Óseas/metabolismo , Enfermedades Óseas/patología , Huesos/citología , Huesos/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Humanos
11.
Proc Natl Acad Sci U S A ; 106(34): 14420-5, 2009 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-19590010

RESUMEN

The role of Notch signaling in cartilage differentiation and maturation in vivo was examined. Conditional Notch pathway gain and loss of function was achieved using a Cre/loxP approach to manipulate Notch signaling in cartilage precursors and chondrocytes of the developing mouse embryo. Conditional overexpression of activated Notch intracellular domain (NICD) in the chondrocyte lineage results in skeletal malformations with decreased cartilage precursor proliferation and inhibited hypertrophic chondrocyte differentiation. Likewise, expression of NICD in cartilage precursors inhibits sclerotome differentiation, resulting in severe axial skeleton abnormalities. Furthermore, conditional loss of Notch signaling via RBP-J gene deletion in the chondrocyte lineage results in increased chondrocyte proliferation and skeletal malformations consistent with the observed increase in hypertrophic chondrocytes. In addition, the Notch pathway inhibits expression of Sox9 and its target genes required for normal chondrogenic cell proliferation and differentiation. Together, our results demonstrate that appropriate Notch pathway signaling is essential for proper chondrocyte progenitor proliferation and for the normal progression of hypertrophic chondrocyte differentiation into bone in the developing appendicular and axial skeletal elements.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular , Condrocitos/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal/fisiología , Animales , Huesos/citología , Huesos/embriología , Huesos/metabolismo , Cartílago/citología , Cartílago/embriología , Cartílago/metabolismo , Diferenciación Celular/genética , Linaje de la Célula , Condrocitos/citología , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Osteoblastos/citología , Osteoblastos/metabolismo , Receptor Notch1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética
12.
Front Mol Biosci ; 9: 1023511, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36339722

RESUMEN

ADAMTS family members control mammalian development and disease, primarily through their function as proteases, by regulation of extracellular matrix composition. Until recently, ADAMTS6 was known as one of the orphan proteinases of the nineteen-member family with a relatively unknown expression pattern and function. Emerging focus on this enzyme has started to uncover these unknowns and revealed a vast importance and requirement of ADAMTS6 in cardiovascular and musculoskeletal development. In addition, ADAMTS6 has been linked to numerous disease settings including several types of cancer. This review summarizes the necessity of ADAMTS6 during development, its role in disease and requirement for essential prospective studies to fully realize its biological implications and potential for therapeutic intervention.

13.
Elife ; 112022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35503090

RESUMEN

The embryonic extracellular matrix (ECM) undergoes transition to mature ECM as development progresses, yet few mechanisms ensuring ECM proteostasis during this period are known. Fibrillin microfibrils are macromolecular ECM complexes serving structural and regulatory roles. In mice, Fbn1 and Fbn2, encoding the major microfibrillar components, are strongly expressed during embryogenesis, but fibrillin-1 is the major component observed in adult tissue microfibrils. Here, analysis of Adamts6 and Adamts10 mutant mouse embryos, lacking these homologous secreted metalloproteases individually and in combination, along with in vitro analysis of microfibrils, measurement of ADAMTS6-fibrillin affinities and N-terminomics discovery of ADAMTS6-cleaved sites, identifies a proteostatic mechanism contributing to postnatal fibrillin-2 reduction and fibrillin-1 dominance. The lack of ADAMTS6, alone and in combination with ADAMTS10 led to excess fibrillin-2 in perichondrium, with impaired skeletal development defined by a drastic reduction of aggrecan and cartilage link protein, impaired BMP signaling in cartilage, and increased GDF5 sequestration in fibrillin-2-rich tissue. Although ADAMTS6 cleaves fibrillin-1 and fibrillin-2 as well as fibronectin, which provides the initial scaffold for microfibril assembly, primacy of the protease-substrate relationship between ADAMTS6 and fibrillin-2 was unequivocally established by reversal of the defects in Adamts6-/- embryos by genetic reduction of Fbn2, but not Fbn1.


Asunto(s)
Proteínas ADAMTS , Microfibrillas , Proteínas ADAMTS/química , Proteínas ADAMTS/genética , Proteínas ADAMTS/metabolismo , Animales , Fibrilina-1/genética , Fibrilina-2/metabolismo , Fibrilinas/metabolismo , Ratones , Microfibrillas/metabolismo , Proteolisis
14.
Methods Mol Biol ; 2043: 207-212, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31463914

RESUMEN

Understanding proteolytic remodeling of extracellular matrix involves the generation of global or conditional knockout mice by homologous recombination in embryonic stem cells or their manipulation through new advanced technologies such as CRISPR-Cas9. These models provide opportunities to understand the roles of ADAMTS genes in skeletogenesis. Whole-mount skeletal preparations are necessary for assessment of the skeletal phenotype. They allow for facile visualization of skeletal patterning, size and shape of skeletal elements, and skeletal structure. This protocol describes the staining of the murine skeleton using Alcian blue to identify cartilage and alizarin red to identify bone.


Asunto(s)
Azul Alcián/química , Antraquinonas/química , Huesos/embriología , Animales , Animales Recién Nacidos , Tipificación del Cuerpo , Huesos/química , Sistemas CRISPR-Cas , Desarrollo Embrionario , Ratones
15.
Methods Mol Biol ; 2043: 173-178, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31463911

RESUMEN

RNA in situ hybridization has an important place in matrix biology, as the only method that allows for in situ discrimination of precise spatial and temporal patterns of gene expression. Whereas immunohistochemistry shows where a matrix protein localizes, ISH identifies the cell of origin. Thus, these methods provide complementary information for insights on the life cycle of matrix molecules, including ADAMTS proteases. This protocol encompasses the staining of tissue sections to reveal expression of the gene of interest.


Asunto(s)
Proteínas ADAMTS/genética , Hibridación in Situ/métodos , Animales , Expresión Génica , Indicadores y Reactivos , Ratones , ARN Mensajero/genética
16.
Methods Mol Biol ; 2043: 261-264, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31463918

RESUMEN

The pericellular matrix (PCM), also known as the pericellular coat or glycocalyx, lies between the plasma membrane and the interstitial extracellular matrix (ECM). It can have a dramatic influence on cell function because of its presence at the interface between the cell and its microenvironment. A common tool used to demonstrate the PCM is the particle exclusion assay in which fixed red blood cells are utilized to outline the boundary of the cell together with its PCM. PCM visualization and quantification provide opportunities to uncover the roles of ADAMTS proteases in PCM remodeling in many cell types and processes.


Asunto(s)
Membrana Celular/ultraestructura , Matriz Extracelular/ultraestructura , Miocitos del Músculo Liso/ultraestructura , Células Cultivadas , Humanos , Masculino , Microscopía Fluorescente , Programas Informáticos
17.
Nat Commun ; 11(1): 1571, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32218432

RESUMEN

Estrogens and progesterone control breast development and carcinogenesis via their cognate receptors expressed in a subset of luminal cells in the mammary epithelium. How they control the extracellular matrix, important to breast physiology and tumorigenesis, remains unclear. Here we report that both hormones induce the secreted protease Adamts18 in myoepithelial cells by controlling Wnt4 expression with consequent paracrine canonical Wnt signaling activation. Adamts18 is required for stem cell activation, has multiple binding partners in the basement membrane and interacts genetically with the basal membrane-specific proteoglycan, Col18a1, pointing to the basement membrane as part of the stem cell niche. In vitro, ADAMTS18 cleaves fibronectin; in vivo, Adamts18 deletion causes increased collagen deposition during puberty, which results in impaired Hippo signaling and reduced Fgfr2 expression both of which control stem cell function. Thus, Adamts18 links luminal hormone receptor signaling to basement membrane remodeling and stem cell activation.


Asunto(s)
Proteínas ADAMTS/metabolismo , Hormonas/farmacología , Glándulas Mamarias Animales/citología , Nicho de Células Madre , Células Madre/metabolismo , Proteínas ADAMTS/deficiencia , Proteínas ADAMTS/genética , Animales , Antígenos CD/metabolismo , Línea Celular , Autorrenovación de las Células/efectos de los fármacos , Epitelio/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Femenino , Fibronectinas/metabolismo , Glicoproteínas/metabolismo , Humanos , Ratones Endogámicos C57BL , Modelos Biológicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Progesterona/metabolismo , Regeneración/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Nicho de Células Madre/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
18.
Matrix Biol ; 77: 117-128, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30201140

RESUMEN

Mutations in the secreted metalloproteinase ADAMTS10 cause recessive Weill-Marchesani syndrome (WMS), comprising ectopia lentis, short stature, brachydactyly, thick skin and cardiac valve anomalies. Dominant WMS caused by FBN1 mutations is clinically similar and affects fibrillin-1 microfibrils, which are a major component of the ocular zonule. ADAMTS10 was previously shown to enhance fibrillin-1 assembly in vitro. Here, Adamts10 null mice were analyzed to determine the impact of ADAMTS10 deficiency on fibrillin microfibrils in vivo. An intragenic lacZ reporter identified widespread Adamts10 expression in the eye, musculoskeletal tissues, vasculature, skin and lung. Adamts10-/- mice had reduced viability on the C57BL/6 background, and although surviving mice were slightly smaller and had stiff skin, they lacked brachydactyly and cardiovascular defects. Ectopia lentis was not observed in Adamts10-/- mice, similar to Fbn1-/- mice, most likely because the mouse zonule contains fibrillin-2 in addition to fibrillin-1. Unexpectedly, in contrast to wild-type eyes, Adamts10-/- zonule fibers were thicker and immunostained strongly with fibrillin-2 antibodies into adulthood, whereas fibrillin-1 staining was reduced. Furthermore, fibrillin-2 staining of hyaloid vasculature remnants persisted post-natally in Adamts10-/- eyes. ADAMTS10 was found to cleave fibrillin-2, providing an explanation for persistence of fibrillin-2 at these sites. Thus, analysis of Adamts10-/- mice led to identification of fibrillin-2 as a novel ADAMTS10 substrate and defined a proteolytic mechanism for clearance of ocular fibrillin-2 at the end of the juvenile period.


Asunto(s)
Proteínas ADAMTS/genética , Ojo/metabolismo , Fibrilina-1/genética , Fibrilina-2/genética , Microfibrillas/metabolismo , Síndrome de Weill-Marchesani/genética , Proteínas ADAMTS/deficiencia , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Modelos Animales de Enfermedad , Ojo/crecimiento & desarrollo , Ojo/patología , Femenino , Fibrilina-1/metabolismo , Fibrilina-2/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Operón Lac , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microfibrillas/patología , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Proteolisis , Transducción de Señal , Piel/crecimiento & desarrollo , Piel/metabolismo , Piel/patología , Síndrome de Weill-Marchesani/metabolismo , Síndrome de Weill-Marchesani/patología
19.
Matrix Biol ; 71-72: 225-239, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29885460

RESUMEN

ADAMTS proteins are a superfamily of 26 secreted molecules comprising two related, but distinct families. ADAMTS proteases are zinc metalloendopeptidases, most of whose substrates are extracellular matrix (ECM) components, whereas ADAMTS-like proteins lack a metalloprotease domain, reside in the ECM and have regulatory roles vis-à-vis ECM assembly and/or ADAMTS activity. Evolutionary conservation and expansion of ADAMTS proteins in mammals is suggestive of crucial embryologic or physiological roles in humans. Indeed, Mendelian disorders or birth defects resulting from naturally occurring ADAMTS2, ADAMTS3, ADAMTS10, ADAMTS13, ADAMTS17, ADAMTS20, ADAMTSL2 and ADAMTSL4 mutations as well as numerous phenotypes identified in genetically engineered mice have revealed ADAMTS participation in major biological pathways. Important roles have been identified in a few acquired conditions. ADAMTS5 is unequivocally implicated in pathogenesis of osteoarthritis via degradation of aggrecan, a major structural proteoglycan in cartilage. ADAMTS7 is strongly associated with coronary artery disease and promotes atherosclerosis. Autoantibodies to ADAMTS13 lead to a platelet coagulopathy, thrombotic thrombocytopenic purpura, which is similar to that resulting from ADAMTS13 mutations. ADAMTS proteins have numerous potential connections to other human disorders that were identified by genome-wide association studies. Here, we review inherited and acquired human disorders in which ADAMTS proteins participate, and discuss progress and prospects in therapeutics.


Asunto(s)
Proteínas ADAMTS/genética , Proteínas ADAMTS/metabolismo , Predisposición Genética a la Enfermedad/genética , Mutación , Animales , Anomalías Congénitas/genética , Enfermedad de la Arteria Coronaria/genética , Matriz Extracelular/metabolismo , Estudio de Asociación del Genoma Completo , Humanos , Familia de Multigenes , Osteoartritis/genética , Púrpura Trombocitopénica Trombótica/genética
20.
JCI Insight ; 3(7)2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29618652

RESUMEN

Heterotopic ossification (HO) is a significant clinical problem with incompletely resolved mechanisms. Here, the secreted metalloproteinases ADAMTS7 and ADAMTS12 are shown to comprise a unique proteoglycan class that protects against a tendency toward HO in mouse hindlimb tendons, menisci, and ligaments. Adamts7 and Adamts12 mRNAs were sparsely expressed in murine forelimbs but strongly coexpressed in hindlimb tendons, skeletal muscle, ligaments, and meniscal fibrocartilage. Adamts7-/- Adamts12-/- mice, but not corresponding single-gene mutants, which demonstrated compensatory upregulation of the intact homolog mRNA, developed progressive HO in these tissues after 4 months of age. Adamts7-/- Adamts12-/- tendons had abnormal collagen fibrils, accompanied by reduced levels of the small leucine-rich proteoglycans (SLRPs) biglycan, fibromodulin, and decorin, which regulate collagen fibrillogenesis. Bgn-/0 Fmod-/- mice are known to have a strikingly similar hindlimb HO to that of Adamts7-/- Adamts12-/- mice, implicating fibromodulin and biglycan reduction as a likely mechanism underlying HO in Adamts7-/- Adamts12-/- mice. Interestingly, degenerated human biceps tendons had reduced ADAMTS7 mRNA compared with healthy biceps tendons, which expressed both ADAMTS7 and ADAMTS12. These results suggest that ADAMTS7 and ADAMTS12 drive an innate pathway protective against hindlimb HO in mice and may be essential for human tendon health.


Asunto(s)
Proteínas ADAMTS/metabolismo , Proteína ADAMTS7/metabolismo , Osificación Heterotópica/patología , Osteoartritis/patología , Tendones/patología , Proteínas ADAMTS/genética , Proteína ADAMTS7/genética , Animales , Línea Celular , Condrocitos , Modelos Animales de Enfermedad , Femenino , Miembro Posterior , Humanos , Masculino , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Osificación Heterotópica/diagnóstico por imagen , Osificación Heterotópica/genética , Tendones/citología , Tendones/diagnóstico por imagen , Tendones/ultraestructura , Microtomografía por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA