Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hepatol Commun ; 8(3)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38358377

RESUMO

BACKGROUND: The prevalence of NAFLD is rapidly increasing. NAFLD can progress to NASH, fibrosis, cirrhosis, and HCC, which will soon become the main causes of liver transplantation. To date, no effective drug for NASH has been approved by the Food and Drug Administration. This is partly due to the lack of reliable human in vitro models. Here, we present a novel human liver spheroid model that can be used to study the mechanisms underlying liver fibrosis formation and degradation. METHODS AND RESULTS: Such spheroids, which contain hepatocytes, stellate cells, KC, and LSECs, spontaneously develop fibrosis that is exacerbated by treatment with free fatty acids. Conditioned medium from activated LSECs caused similar activation of fibrosis in spheroids containing primary human hepatocyte and NPCs, indicating the action of soluble mediators from the LSECs. Spheroids containing LSECs treated with free fatty acids produced tissue inhibitor of metalloproteinases inhibitor 1, a matrix metalloproteinases inhibitor important for fibrosis progression. Tissue inhibitor of metalloproteinases inhibitor 1 knockdown using siRNA led to a reduction in collagen and procollagen accumulation, which could be partially rescued using a potent matrix metalloproteinases inhibitor. Interestingly, tissue inhibitor of metalloproteinases inhibitor 1 was found to be expressed at higher levels, specifically in a subtype of endothelial cells in the pericentral region of human fibrotic livers, than in control livers. CONCLUSION: Potential anti-NASH drugs and compounds were evaluated for their efficacy in reducing collagen accumulation, and we found differences in specificity between spheroids with and without LSECs. This new human NASH model may reveal novel mechanisms for the regulation of liver fibrosis and provide a more appropriate model for screening drugs against NASH.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Hepatopatia Gordurosa não Alcoólica , Estados Unidos , Humanos , Células Endoteliais , Ácidos Graxos não Esterificados , Cirrose Hepática , Pró-Colágeno , Inibidores Teciduais de Metaloproteinases , Metaloproteinases da Matriz , Inibidor Tecidual de Metaloproteinase-1/genética
2.
Science ; 383(6683): eade8064, 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38330107

RESUMO

Penile erection is mediated by the corpora cavernosa, a trabecular-like vascular bed that enlarges upon vasodilation, but its regulation is not completely understood. Here, we show that perivascular fibroblasts in the corpora cavernosa support vasodilation by reducing norepinephrine availability. The effect on penile blood flow depends on the number of fibroblasts, which is regulated by erectile activity. Erection dynamically alters the positional arrangement of fibroblasts, temporarily down-regulating Notch signaling. Inhibition of Notch increases fibroblast numbers and consequently raises penile blood flow. Continuous Notch activation lowers fibroblast numbers and reduces penile blood perfusion. Recurrent erections stimulate fibroblast proliferation and limit vasoconstriction, whereas aging reduces the number of fibroblasts and lowers penile blood flow. Our findings reveal adaptive, erectile activity-dependent modulation of penile blood flow by fibroblasts.


Assuntos
Transportador 1 de Aminoácido Excitatório , Fibroblastos , Ereção Peniana , Pênis , Receptores Notch , Animais , Masculino , Camundongos , Circulação Sanguínea , Transportador 1 de Aminoácido Excitatório/metabolismo , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ereção Peniana/fisiologia , Pênis/irrigação sanguínea , Pênis/fisiologia , Receptores Notch/metabolismo , Transdução de Sinais , Vasoconstrição , Vasodilatação
3.
J Exp Med ; 221(2)2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38117255

RESUMO

In dorsal root ganglia (DRG), macrophages reside close to sensory neurons and have largely been explored in the context of pain, nerve injury, and repair. However, we discovered that most DRG macrophages interact with and monitor the vasculature by sampling macromolecules from the blood. Characterization of the DRG vasculature revealed a specialized endothelial bed that transformed in molecular, structural, and permeability properties along the arteriovenous axis and was covered by macrophage-interacting pericytes and fibroblasts. Macrophage phagocytosis spatially aligned with peak endothelial permeability, a process regulated by enhanced caveolar transcytosis in endothelial cells. Profiling the DRG immune landscape revealed two subsets of perivascular macrophages with distinct transcriptome, turnover, and function. CD163+ macrophages self-maintained locally, specifically participated in vasculature monitoring, displayed distinct responses during peripheral inflammation, and were conserved in mouse and man. Our work provides a molecular explanation for the permeability of the blood-DRG barrier and identifies an unappreciated role of macrophages as integral components of the DRG-neurovascular unit.


Assuntos
Células Endoteliais , Gânglios Espinais , Humanos , Macrófagos , Pericitos , Permeabilidade
4.
J Bone Miner Res ; 37(8): 1545-1561, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35652423

RESUMO

Bone regeneration involves skeletal stem/progenitor cells (SSPCs) recruited from bone marrow, periosteum, and adjacent skeletal muscle. To achieve bone reconstitution after injury, a coordinated cellular and molecular response is required from these cell populations. Here, we show that SSPCs from periosteum and skeletal muscle are enriched in osteochondral progenitors, and more efficiently contribute to endochondral ossification during fracture repair as compared to bone-marrow stromal cells. Single-cell RNA sequencing (RNAseq) analyses of periosteal cells reveal the cellular heterogeneity of periosteum at steady state and in response to bone fracture. Upon fracture, both periosteal and skeletal muscle SSPCs transition from a stem/progenitor to a fibrogenic state prior to chondrogenesis. This common activation pattern in periosteum and skeletal muscle SSPCs is mediated by bone morphogenetic protein (BMP) signaling. Functionally, Bmpr1a gene inactivation in platelet-derived growth factor receptor alpha (Pdgfra)-derived SSPCs impairs bone healing and decreases SSPC proliferation, migration, and osteochondral differentiation. These results uncover a coordinated molecular program driving SSPC activation in periosteum and skeletal muscle toward endochondral ossification during bone regeneration. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Assuntos
Fraturas Ósseas , Periósteo , Diferenciação Celular/fisiologia , Condrogênese , Fraturas Ósseas/metabolismo , Humanos , Músculo Esquelético , Osteogênese/fisiologia , Periósteo/metabolismo , Células-Tronco/metabolismo
5.
Bio Protoc ; 11(15): e4107, 2021 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-34458401

RESUMO

The periosteum covering the outer surface of bone contains skeletal stem/progenitor cells that can efficiently form cartilage and bone during bone repair. Several methods have been described to isolate periosteal cells based on bone scraping and/or enzymatic digestion. Here, we describe an explant culture method to isolate periosteum-derived stem/progenitor cells for subsequent in vitro and in vivo analyses. Periosteal cells (PCs) isolated using this protocol express mesenchymal markers, can be expanded in vitro, and exhibit high regenerative potential after in vivo transplantation at a fracture site, suggesting that this protocol can be employed for PC production to use in new cell-based therapies.

6.
Nat Commun ; 12(1): 2860, 2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-34001878

RESUMO

Bone regenerates by activation of tissue resident stem/progenitor cells, formation of a fibrous callus followed by deposition of cartilage and bone matrices. Here, we show that mesenchymal progenitors residing in skeletal muscle adjacent to bone mediate the initial fibrotic response to bone injury and also participate in cartilage and bone formation. Combined lineage and single-cell RNA sequencing analyses reveal that skeletal muscle mesenchymal progenitors adopt a fibrogenic fate before they engage in chondrogenesis after fracture. In polytrauma, where bone and skeletal muscle are injured, skeletal muscle mesenchymal progenitors exhibit altered fibrogenesis and chondrogenesis. This leads to impaired bone healing, which is due to accumulation of fibrotic tissue originating from skeletal muscle and can be corrected by the anti-fibrotic agent Imatinib. These results elucidate the central role of skeletal muscle in bone regeneration and provide evidence that skeletal muscle can be targeted to prevent persistent callus fibrosis and improve bone healing after musculoskeletal trauma.


Assuntos
Regeneração Óssea/fisiologia , Calo Ósseo/fisiologia , Consolidação da Fratura/fisiologia , Fraturas Ósseas/fisiopatologia , Células-Tronco Mesenquimais/fisiologia , Músculo Esquelético/citologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência/métodos , Osteogênese/fisiologia
7.
Methods Mol Biol ; 2230: 151-165, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33197014

RESUMO

Renal capsule transplantation is a very helpful method to grow embryonic tissues or tumors in a vascular environment, allowing for long-term engraftment and biological analyses. This chapter describes the surgical procedure for the transplantation of embryonic skeletal elements in the renal capsule of adult mice and points out the manipulations that can be applied for assaying the role of angiogenesis during bone development and repair.


Assuntos
Desenvolvimento Ósseo/genética , Transplante de Rim/métodos , Morfogênese/genética , Neovascularização Fisiológica/genética , Túnica Adventícia/crescimento & desenvolvimento , Túnica Adventícia/patologia , Animais , Epitélio/crescimento & desenvolvimento , Epitélio/patologia , Humanos , Rim/crescimento & desenvolvimento , Rim/patologia , Linfangiogênese/genética , Vasos Linfáticos/citologia , Camundongos , Neovascularização Patológica/genética , Organogênese/genética
8.
Stem Cell Reports ; 15(4): 955-967, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32916123

RESUMO

Most organs and tissues in the body, including bone, can repair after an injury due to the activation of endogenous adult stem/progenitor cells to replace the damaged tissue. Inherent dysfunctions of the endogenous stem/progenitor cells in skeletal repair disorders are still poorly understood. Here, we report that Fgfr3Y637C/+ over-activating mutation in Prx1-derived skeletal stem/progenitor cells leads to failure of fracture consolidation. We show that periosteal cells (PCs) carrying the Fgfr3Y637C/+ mutation can engage in osteogenic and chondrogenic lineages, but following transplantation do not undergo terminal chondrocyte hypertrophy and transformation into bone causing pseudarthrosis. Instead, Prx1Cre;Fgfr3Y637C/+ PCs give rise to fibrocartilage and fibrosis. Conversely, wild-type PCs transplanted at the fracture site of Prx1Cre;Fgfr3Y637C/+ mice allow hypertrophic cartilage transition to bone and permit fracture consolidation. The results thus highlight cartilage-to-bone transformation as a necessary step for bone repair and FGFR3 signaling within PCs as a key regulator of this transformation.


Assuntos
Regeneração Óssea , Osso e Ossos/patologia , Cartilagem/patologia , Periósteo/metabolismo , Pseudoartrose/patologia , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Calo Ósseo/patologia , Diferenciação Celular , Consolidação da Fratura , Proteínas de Homeodomínio/metabolismo , Integrases/metabolismo , Camundongos Endogâmicos C57BL , Fenótipo , Tíbia/patologia
9.
Nat Commun ; 9(1): 773, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29472541

RESUMO

Bone regeneration relies on the activation of skeletal stem cells (SSCs) that still remain poorly characterized. Here, we show that periosteum contains SSCs with high bone regenerative potential compared to bone marrow stromal cells/skeletal stem cells (BMSCs) in mice. Although periosteal cells (PCs) and BMSCs are derived from a common embryonic mesenchymal lineage, postnatally PCs exhibit greater clonogenicity, growth and differentiation capacity than BMSCs. During bone repair, PCs can efficiently contribute to cartilage and bone, and integrate long-term after transplantation. Molecular profiling uncovers genes encoding Periostin and other extracellular matrix molecules associated with the enhanced response to injury of PCs. Periostin gene deletion impairs PC functions and fracture consolidation. Periostin-deficient periosteum cannot reconstitute a pool of PCs after injury demonstrating the presence of SSCs within periosteum and the requirement of Periostin in maintaining this pool. Overall our results highlight the importance of analyzing periosteum and PCs to understand bone phenotypes.


Assuntos
Regeneração Óssea , Moléculas de Adesão Celular/metabolismo , Periósteo/citologia , Células-Tronco/metabolismo , Animais , Moléculas de Adesão Celular/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteogênese , Periósteo/metabolismo , Células-Tronco/citologia , Células Estromais/citologia , Células Estromais/metabolismo
10.
Development ; 144(15): 2737-2747, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28694257

RESUMO

Postnatal growth of skeletal muscle largely depends on the expansion and differentiation of resident stem cells, the so-called satellite cells. Here, we demonstrate that postnatal satellite cells express components of the bone morphogenetic protein (BMP) signaling machinery. Overexpression of noggin in postnatal mice (to antagonize BMP ligands), satellite cell-specific knockout of Alk3 (the gene encoding the BMP transmembrane receptor) or overexpression of inhibitory SMAD6 decreased satellite cell proliferation and accretion during myofiber growth, and ultimately retarded muscle growth. Moreover, reduced BMP signaling diminished the adult satellite cell pool. Abrogation of BMP signaling in satellite cell-derived primary myoblasts strongly diminished cell proliferation and upregulated the expression of cell cycle inhibitors p21 and p57 In conclusion, these results show that BMP signaling defines postnatal muscle development by regulating satellite cell-dependent myofiber growth and the generation of the adult muscle stem cell pool.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Animais , Western Blotting , Proteínas Morfogenéticas Ósseas/genética , Proliferação de Células/genética , Proliferação de Células/fisiologia , Células Cultivadas , Feminino , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
11.
Stem Cells ; 33(5): 1501-11, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25594525

RESUMO

Although the importance of muscle in skeletal regeneration is well recognized clinically, the mechanisms by which muscle supports bone repair have remained elusive. Muscle flaps are often used to cover the damaged bone after traumatic injury yet their contribution to bone healing is not known. Here, we show that direct bone-muscle interactions are required for periosteum activation and callus formation, and that muscle grafts provide a source of stem cells for skeletal regeneration. We investigated the role of satellite cells, the muscle stem cells. Satellite cells loss in Pax7(-/-) mice and satellite cell ablation in Pax7(Cre) (ERT) (2/) (+) ;DTA(f/f) mice impaired bone regeneration. Although satellite cells did not contribute as a large source of cells endogenously, they exhibited a potential to contribute to bone repair after transplantation. The fracture healing phenotype in Pax7(Cre) (ERT) (2/) (+) ;DTA(f/f) mice was associated with decreased bone morphogenetic proteins (BMPs), insulin-like growth factor 1, and fibroblast growth factor 2 expression that are normally upregulated in response to fracture in satellite cells. Exogenous rhBMP2 improved bone healing in Pax7(Cre) (ERT) (2/) (+) ;DTA(f/f) mice further supporting the role of satellite cells as a source of growth factors. These results provide the first functional evidence for a direct contribution of muscle to bone regeneration with important clinical implications as it may impact the use of muscle flaps, muscle stem cells, and growth factors in orthopedic applications.


Assuntos
Músculo Esquelético/citologia , Regeneração/fisiologia , Células-Tronco/citologia , Animais , Osso e Ossos/fisiologia , Calo Ósseo/fisiologia , Humanos , Camundongos Endogâmicos C57BL , Mioblastos/citologia , Mioblastos/transplante , Periósteo/fisiologia , Células Satélites de Músculo Esquelético/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...