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








Base de dados
Intervalo de ano de publicação
1.
Biomolecules ; 14(1)2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38254701

RESUMO

Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disorder characterized by episodic yet cumulative heterotopic ossification (HO) of skeletal muscles, tendons, ligaments, and fascia. FOP arises from missense mutations in Activin Receptor type I (ACVR1), a type I bone morphogenetic protein (BMP) receptor. Although initial findings implicated constitutive activity of FOP-variant ACVR1 (ACVR1FOP) and/or hyperactivation by BMPs, it was later shown that HO in FOP requires activation of ACVR1FOP by Activin A. Inhibition of Activin A completely prevents HO in FOP mice, indicating that Activin A is an obligate driver of HO in FOP, and excluding a key role for BMPs in this process. This discovery led to the clinical development of garetosmab, an investigational antibody that blocks Activin A. In a phase 2 trial, garetosmab inhibited new heterotopic bone lesion formation in FOP patients. In contrast, antibodies to ACVR1 activate ACVR1FOP and promote HO in FOP mice. Beyond their potential clinical relevance, these findings have enhanced our understanding of FOP's pathophysiology, leading to the identification of fibroadipogenic progenitors as the cells that form HO, and the discovery of non-signaling complexes between Activin A and wild type ACVR1 and their role in tempering HO, and are also starting to inform biological processes beyond FOP.


Assuntos
Miosite Ossificante , Humanos , Animais , Camundongos , Miosite Ossificante/tratamento farmacológico , Ativinas , Anticorpos Monoclonais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I
2.
Nat Genet ; 55(8): 1277-1287, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37558884

RESUMO

In this study, we leveraged the combined evidence of rare coding variants and common alleles to identify therapeutic targets for osteoporosis. We undertook a large-scale multiancestry exome-wide association study for estimated bone mineral density, which showed that the burden of rare coding alleles in 19 genes was associated with estimated bone mineral density (P < 3.6 × 10-7). These genes were highly enriched for a set of known causal genes for osteoporosis (65-fold; P = 2.5 × 10-5). Exome-wide significant genes had 96-fold increased odds of being the top ranked effector gene at a given GWAS locus (P = 1.8 × 10-10). By integrating proteomics Mendelian randomization evidence, we prioritized CD109 (cluster of differentiation 109) as a gene for which heterozygous loss of function is associated with higher bone density. CRISPR-Cas9 editing of CD109 in SaOS-2 osteoblast-like cell lines showed that partial CD109 knockdown led to increased mineralization. This study demonstrates that the convergence of common and rare variants, proteomics and CRISPR can highlight new bone biology to guide therapeutic development.


Assuntos
Predisposição Genética para Doença , Osteoporose , Humanos , Sequenciamento do Exoma , Osteoporose/genética , Densidade Óssea/genética , Alelos , Fatores de Transcrição/genética , Estudo de Associação Genômica Ampla
3.
Proc Natl Acad Sci U S A ; 120(32): e2309967120, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37523551

RESUMO

Body fat distribution is a heritable risk factor for cardiovascular and metabolic disease. In humans, rare Inhibin beta E (INHBE, activin E) loss-of-function variants are associated with a lower waist-to-hip ratio and protection from type 2 diabetes. Hepatic fatty acid sensing promotes INHBE expression during fasting and in obese individuals, yet it is unclear how the hepatokine activin E governs body shape and energy metabolism. Here, we uncover activin E as a regulator of adipose energy storage. By suppressing ß-agonist-induced lipolysis, activin E promotes fat accumulation and adipocyte hypertrophy and contributes to adipose dysfunction in mice. Mechanistically, we demonstrate that activin E elicits its effect on adipose tissue through ACVR1C, activating SMAD2/3 signaling and suppressing PPARG target genes. Conversely, loss of activin E or ACVR1C in mice increases fat utilization, lowers adiposity, and drives PPARG-regulated gene signatures indicative of healthy adipose function. Our studies identify activin E-ACVR1C as a metabolic rheostat promoting liver-adipose cross talk to restrain excessive fat breakdown and preserve fat mass during prolonged fasting, a mechanism that is maladaptive in obese individuals.


Assuntos
Diabetes Mellitus Tipo 2 , Lipólise , Humanos , Camundongos , Animais , Ativinas/metabolismo , Adiposidade/genética , Diabetes Mellitus Tipo 2/metabolismo , PPAR gama/metabolismo , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo
4.
Nat Commun ; 13(1): 4844, 2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-35999217

RESUMO

Body fat distribution is a major, heritable risk factor for cardiometabolic disease, independent of overall adiposity. Using exome-sequencing in 618,375 individuals (including 160,058 non-Europeans) from the UK, Sweden and Mexico, we identify 16 genes associated with fat distribution at exome-wide significance. We show 6-fold larger effect for fat-distribution associated rare coding variants compared with fine-mapped common alleles, enrichment for genes expressed in adipose tissue and causal genes for partial lipodystrophies, and evidence of sex-dimorphism. We describe an association with favorable fat distribution (p = 1.8 × 10-09), favorable metabolic profile and protection from type 2 diabetes (~28% lower odds; p = 0.004) for heterozygous protein-truncating mutations in INHBE, which encodes a circulating growth factor of the activin family, highly and specifically expressed in hepatocytes. Our results suggest that inhibin ßE is a liver-expressed negative regulator of adipose storage whose blockade may be beneficial in fat distribution-associated metabolic disease.


Assuntos
Diabetes Mellitus Tipo 2 , Subunidades beta de Inibinas/genética , Tecido Adiposo , Adiposidade/genética , Diabetes Mellitus Tipo 2/genética , Exoma/genética , Humanos , Mutação
5.
J Clin Invest ; 132(12)2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35511419

RESUMO

Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disorder whose most debilitating pathology is progressive and cumulative heterotopic ossification (HO) of skeletal muscles, ligaments, tendons, and fascia. FOP is caused by mutations in the type I BMP receptor gene ACVR1, which enable ACVR1 to utilize its natural antagonist, activin A, as an agonistic ligand. The physiological relevance of this property is underscored by the fact that HO in FOP is exquisitely dependent on activation of FOP-mutant ACVR1 by activin A, an effect countered by inhibition of anti-activin A via monoclonal antibody treatment. Hence, we surmised that anti-ACVR1 antibodies that block activation of ACVR1 by ligands should also inhibit HO in FOP and provide an additional therapeutic option for this condition. Therefore, we generated anti-ACVR1 monoclonal antibodies that block ACVR1's activation by its ligands. Surprisingly, in vivo, these anti-ACVR1 antibodies stimulated HO and activated signaling of FOP-mutant ACVR1. This property was restricted to FOP-mutant ACVR1 and resulted from anti-ACVR1 antibody-mediated dimerization of ACVR1. Conversely, wild-type ACVR1 was inhibited by anti-ACVR1 antibodies. These results uncover an additional property of FOP-mutant ACVR1 and indicate that anti-ACVR1 antibodies should not be considered as therapeutics for FOP.


Assuntos
Miosite Ossificante , Ossificação Heterotópica , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/farmacologia , Anticorpos/imunologia , Humanos , Ligantes , Mutação , Miosite Ossificante/genética , Ossificação Heterotópica/genética , Ossificação Heterotópica/patologia , Transdução de Sinais/genética
6.
J Immunol Regen Med ; 142021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34796310

RESUMO

INTRODUCTION: The aim of this study was to investigate the ability of osteoarthritic human chondrocytes to produce articular cartilage (AC) tissues with a reduced inflammatory environment in response to 4 anti-inflammatory nutraceuticals: alpha-tocopherol (Alpha), gallic acid (G), ascorbic acid (AA), and catechin hydrate (C). METHODS: Chondrocytes isolated from patients who underwent total knee arthroplasty surgeries were divided into groups (9 male; mean age, 66.2 ± 3.5 years and 11 female; mean age, 64.2 ± 3.1 years). Cells were cultured based on sex and supplemented with either a negative control (NC) medium or NC plus one of the nutraceuticals at a concentration of 50 µM. At day 21, cultures were characterized histologically, biochemically, and for gene expression of vital markers. RESULTS: At day 21, 62.3% and 66.2% reduction in nitric oxide (NO) content was evident for female and male cells, respectively. G-treatment of female cells resulted in the lowest expression of nitric oxide synthase-2 (NOS2), matrix metalloproteinase-13 (MMP13), and collagen type-10 (COL10). Alpha-treatment of male cells resulted in the lowest expression of NOS2, bone morphogenic protein-2, MMP13, COL10 and tumor necrosis factor alpha induced protein-6 (TNFAIP6) relative to NC. AA and Alpha treatment resulted in the highest glycosaminoglycan (GAG) content for female and male cultures, respectively. CONCLUSION: A sex-dependent response of osteoarthritic chondrocytes to nutraceutical treatment was evident. Our results suggest the use of G for female cells and Alpha for male cells in OA applications seems to be favorable in reducing inflammation and enhancing chondrocytes' ability to form AC tissues.

7.
J Nutr Sci ; 10: e82, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34616553

RESUMO

The in vitro effects of four nutraceuticals, catechin hydrate, gallic acid, α-tocopherol and ascorbic acid, on the ability of human osteoarthritic chondrocytes of two female obese groups to form articular cartilage (AC) tissues and to reduce inflammation were investigated. Group 1 represented thirteen females in the 50-69 years old range, an average weight of 100 kg and an average body mass index (BMI) of 34⋅06 kg/m2. Group 2 was constituted of three females in the 70-80 years old range, an average weight of 75 kg and an average BMI of 31⋅43 kg/m2. The efficacy of nutraceuticals was assessed in monolayer cultures using histological, colorimetric and mRNA gene expression analyses. AC engineered tissues of group 1 produced less total collagen and COL2A1 (38-fold), and higher COL10A1 (2⋅7-fold), MMP13 (50-fold) and NOS2 (15-fold) mRNA levels than those of group 2. In comparison, engineered tissues of group 1 had a significant decrease in NO levels from day 1 to day 21 (2⋅6-fold), as well as higher mRNA levels of FOXO1 (2-fold) and TNFAIP6 (16-fold) compared to group 2. Catechin hydrate decreased NO levels significantly in group 1 (1⋅5-fold) while increasing NO levels significantly in group 2 (3⋅8-fold). No differences from the negative control were observed in the presence of other nutraceuticals for either group. In conclusion, engineered tissues of the younger but heavier patients responded better to nutraceuticals than those from the older but leaner study participants. Finally, cells of group 2 formed better AC tissues with less inflammation and better extracellular matrix than cells of group 1.


Assuntos
Condrócitos/efeitos dos fármacos , Suplementos Nutricionais , Osteoartrite , Idoso , Idoso de 80 Anos ou mais , Ácido Ascórbico/farmacologia , Catequina/farmacologia , Células Cultivadas , Feminino , Ácido Gálico/farmacologia , Humanos , Inflamação , Pessoa de Meia-Idade , Osteoartrite/tratamento farmacológico , RNA Mensageiro , alfa-Tocoferol/farmacologia
8.
Exp Cell Res ; 408(2): 112841, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34563516

RESUMO

Osteoarthritis (OA) patients undergo cartilage degradation and experience painful joint swelling. OA symptoms are caused by inflammatory molecules and the upregulation of catabolic genes leading to the breakdown of cartilage extracellular matrix (ECM). Here, we investigate the effects of gallic acid (GA) and mechanical stretching on the expression of anabolic and catabolic genes and restoring ECM production by osteoarthritic human articular chondrocytes (hAChs) cultured in monolayers. hAChs were seeded onto conventional plates or silicone chambers with or without 100 µM GA. A 5% cyclic tensile strain (CTS) was applied to the silicone chambers and the deposition of collagen and glycosaminoglycan, and gene expressions of collagen types II (COL2A1), XI (COL11A2), I (COL1A1), and X (COL10A1), and matrix metalloproteinases (MMP-1 and MMP-13) as inflammation markers, were quantified. CTS and GA acted synergistically to promote the deposition of collagen and glycosaminoglycan in the ECM by 14- and 7-fold, respectively. Furthermore, the synergistic stimuli selectively upregulated the expression of cartilage-specific proteins, COL11A2 by 7-fold, and COL2A1 by 47-fold, and, in contrast, downregulated the expression of MMP-1 by 2.5-fold and MMP-13 by 125-fold. GA supplementation with CTS is a promising approach for restoring osteoarthritic hAChs ECM production ability making them suitable for complex tissue engineering applications.


Assuntos
Cartilagem Articular/efeitos dos fármacos , Matriz Extracelular/genética , Inflamação/terapia , Exercícios de Alongamento Muscular , Osteoartrite/terapia , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Cadeia alfa 1 do Colágeno Tipo I/genética , Colágeno Tipo II/genética , Colágeno Tipo X/genética , Colágeno Tipo XI/genética , Matriz Extracelular/efeitos dos fármacos , Ácido Gálico/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/genética , Inflamação/patologia , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 13 da Matriz/genética , Osteoartrite/genética , Osteoartrite/patologia
9.
Elife ; 92020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32515349

RESUMO

Activin A functions in BMP signaling in two ways: it either engages ACVR1B to activate Smad2/3 signaling or binds ACVR1 to form a non-signaling complex (NSC). Although the former property has been studied extensively, the roles of the NSC remain unexplored. The genetic disorder fibrodysplasia ossificans progressiva (FOP) provides a unique window into ACVR1/Activin A signaling because in that disease Activin can either signal through FOP-mutant ACVR1 or form NSCs with wild-type ACVR1. To explore the role of the NSC, we generated 'agonist-only' Activin A muteins that activate ACVR1B but cannot form the NSC with ACVR1. Using one of these muteins, we demonstrate that failure to form the NSC in FOP results in more severe disease pathology. These results provide the first evidence for a biological role for the NSC in vivo and pave the way for further exploration of the NSC's physiological role in corresponding knock-in mice.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Ativinas/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Miosite Ossificante/genética , Transdução de Sinais/genética , Receptores de Ativinas Tipo I/genética , Ativinas/genética , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Técnicas de Introdução de Genes , Camundongos , Camundongos Transgênicos , Mutação , Miosite Ossificante/patologia
10.
Proc Natl Acad Sci U S A ; 116(31): 15505-15513, 2019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31315975

RESUMO

TGFß family ligands, which include the TGFßs, BMPs, and activins, signal by forming a ternary complex with type I and type II receptors. For TGFßs and BMPs, structures of ternary complexes have revealed differences in receptor assembly. However, structural information for how activins assemble a ternary receptor complex is lacking. We report the structure of an activin class member, GDF11, in complex with the type II receptor ActRIIB and the type I receptor Alk5. The structure reveals that receptor positioning is similar to the BMP class, with no interreceptor contacts; however, the type I receptor interactions are shifted toward the ligand fingertips and away from the dimer interface. Mutational analysis shows that ligand type I specificity is derived from differences in the fingertips of the ligands that interact with an extended loop specific to Alk4 and Alk5. The study also reveals differences for how TGFß and GDF11 bind to the same type I receptor, Alk5. For GDF11, additional contacts at the fingertip region substitute for the interreceptor interactions that are seen for TGFß, indicating that Alk5 binding to GDF11 is more dependent on direct contacts. In support, we show that a single residue of Alk5 (Phe84), when mutated, abolishes GDF11 signaling, but has little impact on TGFß signaling. The structure of GDF11/ActRIIB/Alk5 shows that, across the TGFß family, different mechanisms regulate type I receptor binding and specificity, providing a molecular explanation for how the activin class accommodates low-affinity type I interactions without the requirement of cooperative receptor interactions.


Assuntos
Ativinas/química , Ativinas/metabolismo , Complexos Multiproteicos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/química , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Humanos , Camundongos , Modelos Moleculares , Complexos Multiproteicos/química , Ratos , Fator de Crescimento Transformador beta/metabolismo
11.
Bone ; 109: 210-217, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28629737

RESUMO

Fibrodysplasia Ossificans Progressiva (FOP) is a rare genetic disorder that presents at birth with only minor patterning defects, but manifests its debilitating pathology early in life with episodic, yet progressive and cumulative, heterotopic ossification (HO) of ligaments, tendons, and a subset of major skeletal muscles. The resulting HO lesions are endochondral in nature, and appear to be linked to inflammatory stimuli arising in association with known injuries, or from inflammation linked to normal tissue repair. FOP is caused by gain-of-function mutations in ACVR1, which encodes a type I BMP receptor. Initial studies on the pathogenic mechanism of FOP-causing mutations in ACVR1 focused on the enhanced function of this receptor in response to certain BMP ligands, or independently of ligands, but did not directly address the fact that HO in FOP is episodic and inflammation-driven. Recently, we and others demonstrated that Activin A is an obligate factor for the initiation of HO in FOP, signaling aberrantly via mutant ACVR1 to transduce osteogenic signals and trigger heterotopic bone formation (Hatsell et al., 2015; Hino et al., 2015). Subsequently, we identified distinct tissue-resident mesenchymal progenitor cells residing in muscles and tendons that recognize Activin A as a pro-osteogenic signal (solely in the context of FOP-causing mutant ACVR1), and give rise to the cartilaginous anlagen that form heterotopic bone (Dey et al., 2016). During the course of these studies, we also found that the activity of FOP-causing ACVR1 mutations does not by itself explain the triggered or inflammatory nature of HO in FOP, suggesting the importance of other, inflammation-introduced, factors or processes. This review presents a synthesis of these findings with a focus on the role of Activin A and inflammation in HO, and lays out perspectives for future research.


Assuntos
Ativinas/metabolismo , Miosite Ossificante/metabolismo , Ossificação Heterotópica/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo , Ativinas/genética , Humanos , Mutação/genética , Miosite Ossificante/genética , Ossificação Heterotópica/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Células-Tronco/metabolismo
12.
J Bone Miner Res ; 32(12): 2489-2499, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28782882

RESUMO

Fibrodysplasia ossificans progressiva (FOP) is a rare autosomal dominant disorder that is characterized by episodic yet cumulative heterotopic ossification (HO) in skeletal muscles, tendons, and ligaments over a patient's lifetime. FOP is caused by missense mutations in the type I bone morphogenetic protein (BMP) receptor ACVR1. We have determined that the formation of heterotopic bone in FOP requires activation of mutant ACVR1 by Activin A, in part by showing that prophylactic inhibition of Activin A blocks HO in a mouse model of FOP. Here we piece together a natural history of developing HO lesions in mouse FOP, and determine where in the continuum of HO Activin A is required, using imaging (T2-MRI, µCT, 18 F-NaF PET/CT, histology) coupled with pharmacologic inhibition of Activin A at different times during the progression of HO. First, we show that expansion of HO lesions comes about through growth and fusion of independent HO events. These events tend to arise within a neighborhood of existing lesions, indicating that already formed HO likely triggers the formation of new events. The process of heterotopic bone expansion appears to be dependent on Activin A because inhibition of this ligand suppresses the growth of nascent HO lesions and stops the emergence of new HO events. Therefore, our results reveal that Activin A is required at least up to the point when nascent HO lesions mineralize and further demonstrate the therapeutic utility of Activin A inhibition in FOP. These results provide evidence for a model where HO is triggered by inflammation but becomes "self-propagating" by a process that requires Activin A. © 2017 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.


Assuntos
Ativinas/metabolismo , Miosite Ossificante/patologia , Ossificação Heterotópica/patologia , Animais , Imageamento por Ressonância Magnética , Camundongos , Miosite Ossificante/diagnóstico por imagem , Ossificação Heterotópica/diagnóstico por imagem , Microtomografia por Raio-X
13.
PLoS One ; 11(2): e0150085, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26910759

RESUMO

Non-bone in vivo micro-CT imaging has many potential applications for preclinical evaluation. Specifically, the in vivo quantification of changes in the vascular network and organ morphology in small animals, associated with the emergence and progression of diseases like bone fracture, inflammation and cancer, would be critical to the development and evaluation of new therapies for the same. However, there are few published papers describing the in vivo vascular imaging in small animals, due to technical challenges, such as low image quality and low vessel contrast in surrounding tissues. These studies have primarily focused on lung, cardiovascular and brain imaging. In vivo vascular imaging of mouse hind limbs has not been reported. We have developed an in vivo CT imaging technique to visualize and quantify vasculature and organ structure in disease models, with the goal of improved quality images. With 1-2 minutes scanning by a high speed in vivo micro-CT scanner (Quantum CT), and injection of a highly efficient contrast agent (Exitron nano 12000), vasculature and organ structure were semi-automatically segmented and quantified via image analysis software (Analyze). Vessels of the head and hind limbs, and organs like the heart, liver, kidneys and spleen were visualized and segmented from density maps. In a mouse model of bone metastasis, neoangiogenesis was observed, and associated changes to vessel morphology were computed, along with associated enlargement of the spleen. The in vivo CT image quality, voxel size down to 20 µm, is sufficient to visualize and quantify mouse vascular morphology. With this technique, in vivo vascular monitoring becomes feasible for the preclinical evaluation of small animal disease models.


Assuntos
Angiografia/métodos , Meios de Contraste/farmacologia , Neoplasias Experimentais , Neovascularização Patológica/diagnóstico por imagem , Microtomografia por Raio-X/métodos , Animais , Camundongos , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/diagnóstico por imagem , Especificidade de Órgãos
14.
Sci Transl Med ; 7(303): 303ra137, 2015 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-26333933

RESUMO

Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disorder characterized by episodically exuberant heterotopic ossification (HO), whereby skeletal muscle is abnormally converted into misplaced, but histologically normal bone. This HO leads to progressive immobility with catastrophic consequences, including death by asphyxiation. FOP results from mutations in the intracellular domain of the type I BMP (bone morphogenetic protein) receptor ACVR1; the most common mutation alters arginine 206 to histidine (ACVR1(R206H)) and has been thought to drive inappropriate bone formation as a result of receptor hyperactivity. We unexpectedly found that this mutation rendered ACVR1 responsive to the activin family of ligands, which generally antagonize BMP signaling through ACVR1 but cannot normally induce bone formation. To test the implications of this finding in vivo, we engineered mice to carry the Acvr1(R206H) mutation. Because mice that constitutively express Acvr1[R206H] die perinatally, we generated a genetically humanized conditional-on knock-in model for this mutation. When Acvr1[R206H] expression was induced, mice developed HO resembling that of FOP; HO could also be triggered by activin A administration in this mouse model of FOP but not in wild-type controls. Finally, HO was blocked by broad-acting BMP blockers, as well as by a fully human antibody specific to activin A. Our results suggest that ACVR1(R206H) causes FOP by gaining responsiveness to the normally antagonistic ligand activin A, demonstrating that this ligand is necessary and sufficient for driving HO in a genetically accurate model of FOP; hence, our human antibody to activin A represents a potential therapeutic approach for FOP.


Assuntos
Receptores de Ativinas Tipo I/genética , Ativinas/metabolismo , Mutação , Miosite Ossificante/genética , Receptores de Ativinas Tipo I/metabolismo , Animais , Camundongos , Camundongos Transgênicos , Ligação Proteica , Proteína 1A de Ligação a Tacrolimo/metabolismo
15.
J Cell Biol ; 189(6): 1027-38, 2010 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-20530211

RESUMO

Rapid plasma membrane resealing is essential for cellular survival. Earlier studies showed that plasma membrane repair requires Ca(2+)-dependent exocytosis of lysosomes and a rapid form of endocytosis that removes membrane lesions. However, the functional relationship between lysosomal exocytosis and the rapid endocytosis that follows membrane injury is unknown. In this study, we show that the lysosomal enzyme acid sphingomyelinase (ASM) is released extracellularly when cells are wounded in the presence of Ca(2+). ASM-deficient cells, including human cells from Niemann-Pick type A (NPA) patients, undergo lysosomal exocytosis after wounding but are defective in injury-dependent endocytosis and plasma membrane repair. Exogenously added recombinant human ASM restores endocytosis and resealing in ASM-depleted cells, suggesting that conversion of plasma membrane sphingomyelin to ceramide by this lysosomal enzyme promotes lesion internalization. These findings reveal a molecular mechanism for restoration of plasma membrane integrity through exocytosis of lysosomes and identify defective plasma membrane repair as a possible component of the severe pathology observed in NPA patients.


Assuntos
Membrana Celular/metabolismo , Endocitose/fisiologia , Exocitose/fisiologia , Esfingomielina Fosfodiesterase/metabolismo , Animais , Linhagem Celular , Membrana Celular/ultraestrutura , Ceramidas/metabolismo , Desipramina/metabolismo , Endossomos/metabolismo , Endossomos/ultraestrutura , Inibidores Enzimáticos/metabolismo , Inativação Gênica , Humanos , Lisossomos/enzimologia , Lisossomos/ultraestrutura , Doenças de Niemann-Pick/metabolismo , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Esfingomielina Fosfodiesterase/genética , Esfingomielinas/metabolismo
16.
Trends Cell Biol ; 18(11): 552-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18848451

RESUMO

Ca(2+) influx through plasma membrane wounds triggers a rapid-repair response that is essential for cell survival. Earlier studies showed that repair requires the exocytosis of intracellular vesicles. Exocytosis was thought to promote resealing by 'patching' the plasma membrane lesion or by facilitating bilayer restoration through reduction in membrane tension. However, cells also rapidly repair lesions created by pore-forming proteins, a form of injury that cannot be resealed solely by exocytosis. Recent studies indicate that, in cells injured by pores or mechanical abrasions, exocytosis is followed by lesion removal through endocytosis. Describing the relationship between wound-induced exocytosis and endocytosis has implications for the understanding of muscular degenerative diseases that are associated with defects in plasma membrane repair.


Assuntos
Cálcio/metabolismo , Membrana Celular/metabolismo , Clatrina/metabolismo , Endocitose , Exocitose , Animais , Sinalização do Cálcio , Cavéolas/metabolismo , Membrana Celular/ultraestrutura , Células HeLa , Humanos , Rim/metabolismo , Rim/ultraestrutura
17.
J Cell Biol ; 180(5): 905-14, 2008 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-18316410

RESUMO

Ca2+ influx through plasma membrane lesions triggers a rapid repair process that was previously shown to require the exocytosis of lysosomal organelles (Reddy, A., E. Caler, and N. Andrews. 2001. Cell. 106:157-169). However, how exocytosis leads to membrane resealing has remained obscure, particularly for stable lesions caused by pore-forming proteins. In this study, we show that Ca2+-dependent resealing after permeabilization with the bacterial toxin streptolysin O (SLO) requires endocytosis via a novel pathway that removes SLO-containing pores from the plasma membrane. We also find that endocytosis is similarly required to repair lesions formed in mechanically wounded cells. Inhibition of lesion endocytosis (by sterol depletion) inhibits repair, whereas enhancement of endocytosis through disruption of the actin cytoskeleton facilitates resealing. Thus, endocytosis promotes wound resealing by removing lesions from the plasma membrane. These findings provide an important new insight into how cells protect themselves not only from mechanical injury but also from microbial toxins and pore-forming proteins produced by the immune system.


Assuntos
Sinalização do Cálcio/fisiologia , Membrana Celular/metabolismo , Endocitose/fisiologia , Células Epiteliais/metabolismo , Proteínas de Membrana/metabolismo , Animais , Toxinas Bacterianas/toxicidade , Cálcio/metabolismo , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/ultraestrutura , Estruturas da Membrana Celular/efeitos dos fármacos , Estruturas da Membrana Celular/metabolismo , Estruturas da Membrana Celular/ultraestrutura , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/ultraestrutura , Células HeLa , Humanos , Imunidade Inata/fisiologia , Ratos , Tempo de Reação/fisiologia , Esteróis/metabolismo , Estreptolisinas/toxicidade , Fatores de Tempo , Cicatrização/fisiologia
18.
Exp Hematol ; 33(1): 42-52, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15661397

RESUMO

OBJECTIVE: Human neutrophil collagenase (HNC) is one of several secondary granule proteins (SGP) expressed late in the myeloid maturation pathway. SGPs are encoded by unlinked and functionally diverse genes that are hypothesized to be coordinately regulated at the transcriptional level and demonstrate uniform dysregulation in leukemic cells. In support of the hypothesis that tissue and stage-specific expression of SGP genes is regulated by shared factor(s), we sought to identify factors responsible for positive regulation of the SGP genes. METHODS: Using 5' deletion analysis, we identified a minimal HNC promoter located within the first 193 bp upstream of the transcription start site. Three CCAAT enhancer binding protein (C/EBP) sites were identified within this region and their functional importance was confirmed by mutational analysis, gel retardation, and oligonucleotide pulldown assays. Using chromatin immunoprecipitation (ChIP), we demonstrated that C/EBPalpha binds to the SGP gene promoters lactoferrin and HNC in nonexpressing cells. Upon induction of maturation, C/EBPalpha binds to these promoters and this binding correlates with the expression of both SGP genes. CONCLUSION: We conclude that in the later stages of myeloid development, SGP genes are coordinately upregulated, and that members of the C/EBP family of transcription factors, in particular C/EBPalpha and C/EBPepsilon, play specific and unique roles in upregulating their expression.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/fisiologia , Regulação da Expressão Gênica/genética , Metaloproteinase 8 da Matriz/genética , Mielopoese/genética , Animais , Sítios de Ligação , Proteína alfa Estimuladora de Ligação a CCAAT , Linhagem Celular , Regulação da Expressão Gênica/fisiologia , Humanos , Lactoferrina/genética , Camundongos , Regiões Promotoras Genéticas , Transfecção , Regulação para Cima
19.
Science ; 304(5676): 1515-8, 2004 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-15178804

RESUMO

Strategies for inhibiting phagolysosome fusion are essential for the intracellular survival and replication of many pathogens. We found that the lysosomal synaptotagmin Syt VII is required for a mechanism that promotes phagolysosomal fusion and limits the intracellular growth of pathogenic bacteria. Syt VII was required for a form of Ca2+-dependent phagolysosome fusion that is analogous to Ca2+-regulated exocytosis of lysosomes, which can be triggered by membrane injury. Bacterial type III secretion systems, which permeabilize membranes and cause Ca2+ influx in mammalian cells, promote lysosomal exocytosis and inhibit intracellular survival in Syt VII +/+ but not -/- cells. Thus, the lysosomal repair response can also protect cells against pathogens that trigger membrane permeabilization.


Assuntos
Bactérias/crescimento & desenvolvimento , Proteínas de Ligação ao Cálcio , Membrana Celular/fisiologia , Glicoproteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Salmonella typhimurium/crescimento & desenvolvimento , Animais , Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células CHO , Cálcio/metabolismo , Células Cultivadas , Cricetinae , Endocitose , Exocitose , Listeria monocytogenes/crescimento & desenvolvimento , Lisossomos/microbiologia , Lisossomos/fisiologia , Macrófagos/microbiologia , Glicoproteínas de Membrana/genética , Camundongos , Mutação , Proteínas do Tecido Nervoso/genética , Permeabilidade , Fagossomos/microbiologia , Fagossomos/fisiologia , Salmonella typhimurium/metabolismo , Sinaptotagminas , Vacúolos/microbiologia , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/crescimento & desenvolvimento
20.
Infect Immun ; 71(8): 4351-60, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12874312

RESUMO

Streptococcus mutans is the principal acidogenic component of dental plaque that demineralizes tooth enamel, leading to dental decay. Cell-associated glucosyltransferases catalyze the sucrose-dependent synthesis of sticky glucan polymers that, together with glucan binding proteins, promote S. mutans adherence to teeth and cell aggregation. We generated an S. mutans Tn916 transposon mutant, GMS315, which is defective in sucrose-dependent adherence and significantly less cariogenic than the UA130 wild-type progenitor in germfree rats. The results of sodium dodecyl sulfate-polyacrylamide gel electrophoresis, Western blotting, and N-terminal sequence analysis confirmed the absence of a 155-kDa glucosyltransferase S (Gtf-S) from GMS315 protein profiles. Mapping of the unique transposon insertion in GMS315 revealed disruption of a putative regulatory region located upstream of gcrR, a gene previously described by Sato et al. that shares significant amino acid identity with other bacterial response regulators (Y. Sato, Y. Yamamoto, and H. Kizaki, FEMS Microbiol. Lett. 186: 187-191, 2000). The gcrR regulator, which we call "tarC," does not align with any of the 13 proposed two-component signal transduction systems derived from in silico analysis of the S. mutans genome, but rather represents one of several orphan response regulators in the genome. The results of Northern hybridization and/or real-time reverse transcription-PCR experiments reveal increased expression of both Gtf-S and glucan binding protein C (GbpC) in a tarC knockout mutant (GMS900), thereby supporting the notion that TarC acts as a negative transcriptional regulator. In addition, we noted that GMS900 has altered biofilm architecture relative to the wild type and is hypocariogenic in germfree rats. Taken collectively, these findings support a role for signal transduction in S. mutans sucrose-dependent adherence and aggregation and implicate TarC as a potential target for controlling S. mutans-induced cariogenesis.


Assuntos
Aderência Bacteriana/fisiologia , Cárie Dentária/etiologia , Streptococcus mutans/fisiologia , Streptococcus mutans/patogenicidade , Sequência de Aminoácidos , Animais , Aderência Bacteriana/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Sequência de Bases , Biofilmes , DNA Bacteriano/genética , Cárie Dentária/microbiologia , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Vida Livre de Germes , Glucosiltransferases/genética , Glucosiltransferases/fisiologia , Humanos , Microscopia Eletrônica de Varredura , Dados de Sequência Molecular , Mutagênese Insercional , Ratos , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Streptococcus mutans/genética , Sacarose/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA