Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Orthop Res ; 42(5): 950-960, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-37975633

RESUMO

Collagen V (Col5) is a quantitatively minor component of collagen fibrils comprising tendon, however, plays a crucial role in regulation of development and dynamic healing processes. Clinically, patients with COL5a1 haploinsufficiency, known as classic Ehlers-Danlos Syndrome (cEDS), present with hyperextensible skin, joint instability and laxity, with females more likely to be affected. Previous studies in Col5-deficient mice indicated that reduced Col5a1 expression leads to a reduction in stiffness, fibril deposition, and altered fibril structure. Additionally, Col5-deficient male tendons demonstrated altered healing compared to wild-type tendons, however female mice have not yet been studied utilizing this model. Along with clinical differences between sexes in cEDS patient populations, differences in hormone physiology may be a factor influencing tendon health. Therefore, the objective of this study was to utilize a Col5a1+/ - female mouse model, to determine the effect of Col5 on tendon cell morphology, cell density, tissue composition, and mechanical properties throughout healing. We hypothesized that reduction in Col5 expression would result in an abnormal wound matrix post-injury, resulting in reduced mechanical properties compared to normal tendons. Following patellar tendon surgery, mice were euthanized at 1, 3, and 6-week post-injury. Col5-deficient tendons demonstrated altered and decreased healing compared to WT tendons. The lack of resolution in cellularity by 6-week post-injury in Col5-deficient tendons influenced the decreased mechanical properties. Stiffness did not increase post-injury in Col5-deficient mice, and collagen fiber realignment was delayed during mechanical loading. Therefore, increased Col5a1 expression post-injury is necessary to re-establish matrix engagement and cellularity throughout tendon healing.


Assuntos
Síndrome de Ehlers-Danlos , Ligamento Patelar , Camundongos , Humanos , Masculino , Animais , Feminino , Haploinsuficiência , Colágeno/metabolismo , Tendões/metabolismo , Contagem de Células
2.
Nat Cell Biol ; 23(7): 771-781, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34239060

RESUMO

Tissue turnover requires activation and lineage commitment of tissue-resident stem cells (SCs). These processes are impacted by ageing, but the mechanisms remain unclear. Here, we addressed the mechanisms of ageing in murine hair follicle SCs (HFSCs) and observed a widespread reduction in chromatin accessibility in aged HFSCs, particularly at key self-renewal and differentiation genes, characterized by bivalent promoters occupied by active and repressive chromatin marks. Consistent with this, aged HFSCs showed reduced ability to activate bivalent genes for efficient self-renewal and differentiation. These defects were niche dependent as the transplantation of aged HFSCs into young recipients or synthetic niches restored SC functions. Mechanistically, the aged HFSC niche displayed widespread alterations in extracellular matrix composition and mechanics, resulting in mechanical stress and concomitant transcriptional repression to silence promoters. As a consequence, increasing basement membrane stiffness recapitulated age-related SC changes. These data identify niche mechanics as a central regulator of chromatin state, which, when altered, leads to age-dependent SC exhaustion.


Assuntos
Diferenciação Celular , Autorrenovação Celular , Senescência Celular , Montagem e Desmontagem da Cromatina , Folículo Piloso/fisiologia , Regiões Promotoras Genéticas , Nicho de Células-Tronco , Células-Tronco/fisiologia , Animais , Diferenciação Celular/genética , Linhagem da Célula , Autorrenovação Celular/genética , Células Cultivadas , Senescência Celular/genética , Matriz Extracelular/fisiologia , Inativação Gênica , Folículo Piloso/citologia , Folículo Piloso/metabolismo , Masculino , Mecanotransdução Celular , Camundongos Endogâmicos C57BL , Camundongos Knockout , Envelhecimento da Pele , Células-Tronco/metabolismo , Estresse Mecânico , Transcrição Gênica
3.
Cell Rep ; 31(13): 107818, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32610121

RESUMO

Obesity is characterized by adipose tissue inflammation. Because proteoglycans regulate inflammation, here we investigate their role in adipose tissue inflammation in obesity. We find that adipose tissue versican and biglycan increase in obesity. Versican is produced mainly by adipocytes and biglycan by adipose tissue macrophages. Both proteoglycans are also present in adipose tissue from obese human subjects undergoing gastric bypass surgery. Deletion of adipocyte-specific versican or macrophage-specific biglycan in mice reduces macrophage accumulation and chemokine and cytokine expression, although only adipocyte-specific versican deletion leads to sustained improvement in glucose tolerance. Macrophage-derived biglycan activates inflammatory genes in adipocytes. Versican expression increases in cultured adipocytes exposed to excess glucose, and adipocyte-conditioned medium stimulates inflammation in resident peritoneal macrophages, in part because of a versican breakdown product, versikine. These findings provide insights into the role of adipocyte- and macrophage-derived proteoglycans in adipose tissue inflammation in obesity.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/patologia , Biglicano/metabolismo , Inflamação/patologia , Macrófagos/metabolismo , Obesidade/patologia , Versicanas/metabolismo , Células 3T3-L1 , Animais , Medula Óssea/metabolismo , Dieta Hiperlipídica , Feminino , Teste de Tolerância a Glucose , Humanos , Hipertrofia , Resistência à Insulina , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Pessoa de Meia-Idade , Omento/metabolismo , Especificidade de Órgãos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Gordura Subcutânea/patologia , Versicanas/genética
4.
Mol Vis ; 25: 415-426, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31523119

RESUMO

Purpose: Maintenance of a transparent corneal stroma is imperative for proper vision. The corneal stroma is composed of primarily collagen fibrils, small leucine-rich proteoglycans (SLRPs), as well as sparsely distributed cells called keratocytes. The lattice arrangement and spacing of the collagen fibrils that allows for transparency may be disrupted due to genetic mutations and injuries. The purpose of this study is to examine the therapeutic efficacy of human umbilical cord mesenchymal stem/stromal cells (UMSCs) in treating congenital and acquired corneal opacity associated with the loss of collagen V. Methods: Experimental mice, i.e., wild-type, Col5a1f/f and Kera-Cre/Col5a1f/f (Col5a1∆st/∆st , collagen V null in the corneal stroma) mice in a C57BL/6J genetic background, were subjected to a lamellar keratectomy, and treated with or without UMSC (104 cells/cornea) transplantation via an intrastromal injection or a fibrin plug. In vivo Heidelberg retinal tomograph (HRT II) confocal microscopy, second harmonic generated (SHG) confocal microscopy, histology, and immunofluorescence microscopy were used to assess the corneal transparency of the regenerated corneas. Results: Col5a1∆st/∆st mice display a cloudy cornea phenotype that is ameliorated following intrastromal transplantation of UMSCs. Loss of collagen V in Col5a1∆st/∆st corneas augments the formation of cornea scarring following the keratectomy. UMSC transplantation with a fibrin plug improves the healing of injured corneas and regeneration of transparent corneas, as determined with in vivo HRT II confocal microscopy. Second harmonic confocal microscopy revealed the improved collagen fibril lamellar architecture in the UMSC-transplanted cornea in comparison to the control keratectomized corneas. Conclusions: UMSC transplantation was successful in recovering some corneal transparency in injured corneas of wild-type, Col5a1f/f and Col5a1∆st/∆st mice. The production of collagen V by transplanted UMSCs may account for the regeneration of corneal transparency, as exemplified by better collagen fiber organization, as revealed with SHG signals.


Assuntos
Opacidade da Córnea/congênito , Opacidade da Córnea/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Animais , Colágeno Tipo V/metabolismo , Opacidade da Córnea/patologia , Substância Própria/patologia , Colágenos Fibrilares/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Resultado do Tratamento , Cordão Umbilical/citologia
5.
Matrix Biol ; 82: 38-53, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30738849

RESUMO

Geleophysic dysplasia is a rare, frequently lethal condition characterized by severe short stature with progressive joint contractures, cardiac, pulmonary, and skin anomalies. Geleophysic dysplasia results from dominant fibrillin-1 (FBN1) or recessive ADAMTSL2 mutations, suggesting a functional link between ADAMTSL2 and fibrillin microfibrils. Mice lacking ADAMTSL2 die at birth, which has precluded analysis of postnatal limb development and mechanisms underlying the skeletal anomalies of geleophysic dysplasia. Here, detailed expression analysis of Adamtsl2 using an intragenic lacZ reporter shows strong Adamtsl2 expression in limb tendons. Expression in developing and growing bones is present in regions that are destined to become articular cartilage but is absent in growth plate cartilage. Consistent with strong tendon expression, Adamtsl2 conditional deletion in limb mesenchyme using Prx1-Cre led to tendon anomalies, albeit with normal collagen fibrils, and distal limb shortening, providing a mouse model for geleophysic dysplasia. Unexpectedly, conditional Adamtsl2 deletion using Scx-Cre, a tendon-specific Cre-deleter strain, which does not delete in cartilage, also impaired skeletal growth. Recombinant ADAMTSL2 is shown here to colocalize with fibrillin microfibrils in vitro, and enhanced staining of fibrillin-1 microfibrils was observed in Prx1-Cre Adamtsl2 tendons. The findings show that ADAMTSL2 specifically regulates microfibril assembly in tendons and that proper microfibril composition in tendons is necessary for tendon growth. We speculate that reduced bone growth in geleophysic dysplasia may result from external tethering by short tendons rather than intrinsic growth plate anomalies. Taken together with previous work, we suggest that GD results from abnormal microfibril assembly in tissues, and that ADAMTSL2 may limit the assembly of fibrillin microfibrils.


Assuntos
Proteínas ADAMTS/genética , Doenças do Desenvolvimento Ósseo/genética , Extremidades/crescimento & desenvolvimento , Deleção de Genes , Deformidades Congênitas dos Membros/genética , Tendões/crescimento & desenvolvimento , Proteínas ADAMTS/metabolismo , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Fibrilina-1/metabolismo , Fibrilina-2/metabolismo , Fibrilinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Especificidade de Órgãos , Tendões/metabolismo
7.
Sci Rep ; 7: 41286, 2017 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-28128337

RESUMO

The cornea is the transparent outermost surface of the eye, consisting of a stratified epithelium, a collagenous stroma and an innermost single-cell layered endothelium and providing 2/3 of the refractive power of the eye. Multiple diseases of the cornea arise from genetic defects where the ultimate phenotype can be influenced by cross talk between the cell types and the extracellular matrix. Cell culture modeling of diseases can benefit from cornea organoids that include multiple corneal cell types and extracellular matrices. Here we present human iPS cell-derived organoids through sequential rounds of differentiation programs. These organoids share features of the developing cornea, harboring three distinct cell types with expression of key epithelial, stromal and endothelial cell markers. Cornea organoid cultures provide a powerful 3D model system for investigating corneal developmental processes and their disruptions in diseased conditions.


Assuntos
Diferenciação Celular/genética , Córnea/crescimento & desenvolvimento , Células-Tronco Pluripotentes Induzidas/citologia , Organoides/crescimento & desenvolvimento , Técnicas de Cultura de Células , Córnea/citologia , Células Endoteliais/citologia , Células Epiteliais/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Organoides/citologia , Células Estromais/citologia
8.
Matrix Biol ; 56: 114-131, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27363389

RESUMO

Closely related extracellular metalloproteinases bone morphogenetic protein 1 (BMP1) and mammalian Tolloid-like 1 (mTLL1) are co-expressed in various tissues and have been suggested to have overlapping roles in the biosynthetic processing of extracellular matrix components. Early lethality of mice null for the BMP1 gene Bmp1 or the mTLL1 gene Tll1 has impaired in vivo studies of these proteinases. To overcome issues of early lethality and functional redundancy we developed the novel BTKO mouse strain, with floxed Bmp1 and Tll1 alleles, for induction of postnatal, simultaneous ablation of the two genes. We previously showed these mice to have a skeletal phenotype that includes elements of osteogenesis imperfecta (OI), osteomalacia, and deficient osteocyte maturation, observations validated by the finding of BMP1 mutations in a subset of human patients with OI-like phenotypes. However, the roles of BMP1-like proteinase in non-skeletal tissues have yet to be explored, despite the supposed importance of putative substrates of these proteinases in such tissues. Here, we employ BTKO mice to investigate potential roles for these proteinases in skin. Loss of BMP1-like proteinase activity is shown to result in markedly thinned and fragile skin with unusually densely packed collagen fibrils and delayed wound healing. We demonstrate deficits in the processing of collagens I and III, decorin, biglycan, and laminin 332 in skin, which indicate mechanisms whereby BMP1-like proteinases affect the biology of this tissue. In contrast, lack of effects on collagen VII processing or deposition indicates this putative substrate to be biosynthetically processed by non-BMP1-like proteinases.


Assuntos
Proteína Morfogenética Óssea 1/genética , Derme/enzimologia , Metaloproteases Semelhantes a Toloide/genética , Animais , Biglicano/metabolismo , Proteína Morfogenética Óssea 1/metabolismo , Células Cultivadas , Decorina/metabolismo , Derme/citologia , Técnicas de Inativação de Genes , Masculino , Camundongos Transgênicos , Reepitelização , Metaloproteases Semelhantes a Toloide/metabolismo
9.
J Orthop Res ; 34(7): 1256-63, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26704368

RESUMO

Injury adversely impacts the structure and mechanical properties of a tendon, thus causing pain and disability. Previously, we demonstrated that patellar tendons in mature (P150) and aged (P300) mice do not recover original functionality, even 6 weeks after injury, and that uninjured geriatric tendons (P570) are functionally inferior to uninjured mature tendons. In this study, we hypothesized that the repair response in injured geriatric mice would be further compromised, thus undermining patellar tendon function post-injury. Patellar tendons from wild-type mice were injured at 540 days. At 3 and 6 weeks post-surgery, structural, mechanical, and biochemical analyses were performed and compared to uninjured controls. Mechanical properties of geriatric tendons failed to improve after injury. When compared to mature and aged tendons post-injury, it was determined that at no age was there a suitable repair response. In previous studies, we were able to associate the absence of SLRPs with phenotypic changes both early and late in repair. Here we found that SLRPs were significantly decreased after injury, thus offering a possible explanation for why geriatric tendons were unable to mount an adequate repair response. Thus, we conclude that regardless of age after maturity, tendon healing ultimately results in a substandard outcome. © 2015 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1256-1263, 2016.


Assuntos
Envelhecimento/fisiologia , Ligamento Patelar/lesões , Regeneração , Traumatismos dos Tendões/fisiopatologia , Animais , Feminino , Camundongos Endogâmicos C57BL , Ligamento Patelar/metabolismo , Ligamento Patelar/patologia , Ligamento Patelar/fisiopatologia , Proteoglicanos Pequenos Ricos em Leucina/metabolismo , Traumatismos dos Tendões/metabolismo , Traumatismos dos Tendões/patologia
10.
Sci Transl Med ; 6(266): 266ra172, 2014 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-25504883

RESUMO

Conventional allograft therapy for corneal scarring is widespread and successful, but donor tissue is not universally available, and some grafts fail owing to rejection and complications such as endothelial failure. We investigated direct treatment of corneal scarring using autologous stem cells, a therapy that, if successful, could reduce the need for corneal grafts. Mesenchymal cells were expanded from small superficial, clinically replicable limbal biopsies of human cadaveric corneo-scleral rims. Limbal biopsy-derived stromal cells (LBSCs) expanded rapidly in media containing human serum, were highly clonogenic, and could generate spheres expressing stem cell genes (ABCG2, Nestin, NGFR, Oct4, PAX6, and Sox2). Human LBSCs differentiated into keratocytes expressing characteristic marker genes (ALDH3A1, AQP1, KERA, and PTGDS) and organized a thick lamellar stroma-like tissue containing aligned collagen and keratan sulfate proteoglycans when cultured on aligned nanofiber substrata. When engrafted into mouse corneal wounds, LBSCs prevented formation of light-scattering scar tissue containing fibrotic matrix components. The presence of LBSCs induced regeneration of ablated stroma with tissue exhibiting lamellar structure and collagen organization indistinguishable from that of native tissue. Because the limbus can be easily biopsied from either eye of an affected individual and LBSCs capable of corneal stromal remodeling can be expanded under xeno-free autologous conditions, these cells present a potential for autologous stem cell-based treatment of corneal stromal blindness.


Assuntos
Córnea/patologia , Transplante de Córnea/métodos , Transplante de Células-Tronco , Células-Tronco/citologia , Células Estromais/citologia , Adulto , Animais , Biópsia , Diferenciação Celular , Cicatriz/prevenção & controle , Colágeno/química , Córnea/metabolismo , Meios de Cultura/química , Feminino , Fibroblastos/citologia , Regulação da Expressão Gênica , Humanos , Sulfato de Queratano/química , Queratinócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteoglicanas/química
11.
Hum Mol Genet ; 23(12): 3085-101, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24419319

RESUMO

Osteogenesis imperfecta (OI), or brittle bone disease, is most often caused by dominant mutations in the collagen I genes COL1A1/COL1A2, whereas rarer recessive OI is often caused by mutations in genes encoding collagen I-interacting proteins. Recently, mutations in the gene for the proteinase bone morphogenetic 1 (BMP1) were reported in two recessive OI families. BMP1 and the closely related proteinase mammalian tolloid-like 1 (mTLL1) are co-expressed in various tissues, including bone, and have overlapping activities that include biosynthetic processing of procollagen precursors into mature collagen monomers. However, early lethality of Bmp1- and Tll1-null mice has precluded use of such models for careful study of in vivo roles of their protein products. Here we employ novel mouse strains with floxed Bmp1 and Tll1 alleles to induce postnatal, simultaneous ablation of the two genes, thus avoiding barriers of Bmp1(-/-) and Tll1(-/-) lethality and issues of functional redundancy. Bones of the conditionally null mice are dramatically weakened and brittle, with spontaneous fractures-defining features of OI. Additional skeletal features include osteomalacia, thinned/porous cortical bone, reduced processing of procollagen and dentin matrix protein 1, remarkably high bone turnover and defective osteocyte maturation that is accompanied by decreased expression of the osteocyte marker and Wnt-signaling inhibitor sclerostin, and by marked induction of canonical Wnt signaling. The novel animal model presented here provides new opportunities for in-depth analyses of in vivo roles of BMP1-like proteinases in bone and other tissues, and for their roles, and for possible therapeutic interventions, in OI.


Assuntos
Proteína Morfogenética Óssea 1/genética , Fêmur/patologia , Técnicas de Silenciamento de Genes/métodos , Osteogênese Imperfeita/patologia , Metaloproteases Semelhantes a Toloide/genética , Animais , Proteína Morfogenética Óssea 1/metabolismo , Modelos Animais de Doenças , Fêmur/ultraestrutura , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Osteogênese Imperfeita/genética , Metaloproteases Semelhantes a Toloide/metabolismo
12.
Matrix Biol ; 35: 232-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24157578

RESUMO

Recent studies have demonstrated that the small leucine-rich proteoglycans (SLRPs) biglycan and decorin impact tendon development, aging and healing in mature mice. However, despite the increased risk of tendon injury in the elderly, the role of SLRPs in tendon repair has not been investigated in aged animals. Therefore, our objective was to elucidate the influences of bigylcan and decorin on tendon healing in aged mice to relate our findings to previous work in mature mice. Since the processes of aging and healing are known to interact, our hypothesis was that aging mediates the role of biglycan and decorin on tendon healing. Patellar tendons from wild-type, biglycan-null and decorin-null mice were injured at 270 days using an established model. At 3 and 6 weeks post-surgery, structural, mechanical and biochemical analyses were performed and compared to uninjured controls. Early stage healing was inferior in biglycan-null and decorin-null mice as compared to wild type. However, tendons of all genotypes failed to exhibit improved mechanical properties between 3 and 6 weeks post-injury. In contrast, in a previous investigation of tendon healing in mature (i.e., 120 day-old) mice, only biglycan-null mice were deficient in early stage healing while decorin-null mice were deficient in late-stage healing. These results confirm that the impact of SLRPs on tendon healing is mediated by age and could inform future age-specific therapies for enhancing tendon healing.


Assuntos
Envelhecimento/fisiologia , Biglicano/metabolismo , Decorina/metabolismo , Traumatismos dos Tendões/metabolismo , Cicatrização/fisiologia , Envelhecimento/patologia , Animais , Biglicano/genética , Fenômenos Biomecânicos , Primers do DNA/genética , Decorina/genética , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Ligamento Patelar/lesões , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Cicatrização/genética
13.
J Biomech Eng ; 135(4): 041005, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24231900

RESUMO

While useful models have been proposed to predict the mechanical impact of damage in tendon and other soft tissues, the applicability of these models for describing in vivo injury and age-related degeneration has not been investigated. Therefore, the objective of this study was to develop and validate a simple damage model to predict mechanical alterations in mouse patellar tendons after aging, injury, or healing. To characterize baseline properties, uninjured controls at age 150 days were cyclically loaded across three strain levels and five frequencies. For comparison, damage was induced in mature (120 day-old) mice through either injury or aging. Injured mice were sacrificed at three or six weeks after surgery, while aged mice were sacrificed at either 300 or 570 days old. Changes in mechanical properties (relative to baseline) in the three week post-injury group were assessed and used to develop an empirical damage model based on a simple damage parameter related to the equilibrium stress at a prescribed strain (6%). From the derived model, the viscoelastic properties of the 300 day-old, 570 day-old, and six week post-injury groups were accurately predicted. Across testing conditions, nearly all correlations between predicted and measured parameters were statistically significant and coefficients of determination ranged from R² = 0.25 to 0.97. Results suggest that the proposed damage model could exploit simple in vivo mechanical measurements to predict how an injured or aged tendon will respond to complex physiological loading regimens.


Assuntos
Envelhecimento , Simulação por Computador , Ligamento Patelar/lesões , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Ligamento Patelar/fisiologia , Estresse Mecânico
14.
J Biol Chem ; 288(20): 14320-14331, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23564457

RESUMO

Collagen VI is a ubiquitously expressed extracellular microfibrillar protein. Its most common molecular form is composed of the α1(VI), α2(VI), and α3(VI) collagen α chains encoded by the COL6A1, COL6A2, and COL6A3 genes, respectively. Mutations in any of the three collagen VI genes cause congenital muscular dystrophy types Bethlem and Ullrich as well as intermediate phenotypes characterized by muscle weakness and connective tissue abnormalities. The α3(VI) collagen α chain has much larger N- and C-globular domains than the other two chains. Its most C-terminal domain can be cleaved off after assembly into microfibrils, and the cleavage product has been implicated in tumor angiogenesis and progression. Here we characterize a Col6a3 mutant mouse that expresses a very low level of a non-functional α3(VI) collagen chain. The mutant mice are deficient in extracellular collagen VI microfibrils and exhibit myopathic features, including decreased muscle mass and contractile force. Ultrastructurally abnormal collagen fibrils were observed in tendon, but not cornea, of the mutant mice, indicating a distinct tissue-specific effect of collagen VI on collagen I fibrillogenesis. Overall, the mice lacking normal α3(VI) collagen chains displayed mild musculoskeletal phenotypes similar to mice deficient in the α1(VI) collagen α chain, suggesting that the cleavage product of the α3(VI) collagen does not elicit essential functions in normal growth and development. The Col6a3 mouse mutant lacking functional α3(VI) collagen chains thus serves as an animal model for COL6A3-related muscular dystrophy.


Assuntos
Colágeno Tipo VI/deficiência , Colágeno Tipo VI/genética , Músculo Esquelético/metabolismo , Tendões/metabolismo , Animais , Colágeno Tipo VI/fisiologia , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Genótipo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microfibrilas/metabolismo , Músculo Esquelético/fisiopatologia , Mutação , Fenótipo , Tendões/fisiopatologia
15.
Tissue Eng Part A ; 19(1-2): 199-210, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22871316

RESUMO

Specific niches may affect how cells from different regions contribute to tendon biology, particularly in regard to the healing of certain tendinopathies. The objectives of this study are to determine whether distinct subpopulations of stem/progenitor cells are found within the tendon proper and the epi- and paratenon, the peritenon, as well as to characterize these stem/progenitor cell populations. In this study, we hypothesized that tendon stem/progenitor cells exist in each region, that these populations possess distinct features, and that these populations while multipotent could have differing potentials. To test this hypothesis, stem/progenitor cells were isolated and characterized from the peritenon and tendon proper of mouse Achilles tendons. Colony-forming unit and multipotency assays, as well as flow cytometry, and real-time quantitative polymerase chain reaction analyses of stem cell markers were performed. Significantly, more stem/progenitor cell colonies were observed from cells derived from the tendon proper relative to the peritenon. Analysis of surface markers for stem/progenitor cells from both regions indicated that they were Sca1(+) (stem cell marker), Cd90(+) and Cd44(+) (fibroblast markers), Cd18(-) (leukocyte marker), Cd34(-) (hematopoietic and vascular marker), and Cd133(-) (perivascular marker). Tendon proper stem/progenitor cells had increased expression levels for tenomodulin (Tnmd) and scleraxis (Scx), indicative of enrichment of stem/progenitor cells of a tendon origin. In contrast, cells of the peritenon demonstrated relative increases in the vascular (endomucin) and pericyte (Cd133) markers relative to cells from the tendon proper. Stem/progenitor cells from both regions were multipotent (adipogenic, chondrogenic, osteogenic, and tenogenic). These findings demonstrated that different progenitor populations exist within discrete niches of the Achilles tendon-tendon proper versus peritenon. Overall, these data support the hypothesis that the progenitor pools from both regions have distinct properties and contain enriched progenitor subpopulations of different origins. Moreover, in considering their roles in tendon healing more broadly, they are potential cell sources that may differentially contribute to intrinsic and extrinsic tendon repair mechanisms. That is, intrinsic repair may require a progenitor class with predominant tendon marker expression, while extrinsic repair may involve a progenitor class recruited from perivascular cells of the peritenon.


Assuntos
Tendão do Calcâneo/citologia , Células-Tronco/classificação , Células-Tronco/citologia , Tendão do Calcâneo/metabolismo , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/metabolismo
16.
Stem Cells ; 27(7): 1635-42, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19544455

RESUMO

Corneal scarring from trauma and inflammation disrupts vision for millions worldwide, but corneal transplantation, the primary therapy for corneal blindness, is unavailable to many affected individuals. In this study, stem cells isolated from adult human corneal stroma were examined for the ability to correct stromal opacity in a murine model by direct injection of cells into the corneal stroma. In wild-type mice, injected human stem cells remained viable for months without fusing with host cells or eliciting an immune T-cell response. Human corneal-specific extracellular matrix, including the proteoglycans lumican and keratocan, accumulated in the treated corneas. Lumican-null mice have corneal opacity similar to that of scar tissue as a result of disruption of stromal collagen organization. After injection with human stromal stem cells, stromal thickness and collagen fibril defects in these mice were restored to that of normal mice. Corneal transparency in the treated mice was indistinguishable from that of wild-type mice. These results support the immune privilege of adult stem cells and the ability of stem cell therapy to regenerate tissue in a manner analogous to organogenesis and clearly different from that of normal wound healing. The results suggest that cell-based therapy can be an effective approach to treatment of human corneal blindness.


Assuntos
Doenças da Córnea/terapia , Transplante de Células-Tronco/métodos , Transplante Heterólogo/métodos , Animais , Western Blotting , Células Cultivadas , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Doenças da Córnea/metabolismo , Doenças da Córnea/patologia , Substância Própria/metabolismo , Substância Própria/patologia , Substância Própria/ultraestrutura , Feminino , Citometria de Fluxo , Sulfato de Queratano/metabolismo , Lumicana , Masculino , Camundongos , Microscopia Eletrônica de Transmissão , Proteoglicanas/metabolismo
17.
J Biol Chem ; 283(20): 13736-44, 2008 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-18319262

RESUMO

Fhit protein is lost in most cancers, its restoration suppresses tumorigenicity, and virus-mediated FHIT gene therapy induces apoptosis and suppresses tumors in preclinical models. We have used protein cross-linking and proteomics methods to characterize a Fhit protein complex involved in triggering Fhit-mediated apoptosis. The complex includes Hsp60 and Hsp10 that mediate Fhit stability and may affect import into mitochondria, where it interacts with ferredoxin reductase, responsible for transferring electrons from NADPH to cytochrome P450 via ferredoxin. Viral-mediated Fhit restoration increases production of intracellular reactive oxygen species, followed by increased apoptosis of lung cancer cells under oxidative stress conditions; conversely, Fhit-negative cells escape apoptosis, carrying serious oxidative DNA damage that may contribute to an increased mutation rate. Characterization of Fhit interacting proteins has identified direct effectors of the Fhit-mediated apoptotic pathway that is lost in most cancers through loss of Fhit.


Assuntos
Hidrolases Anidrido Ácido/metabolismo , Apoptose , Ferredoxina-Nitrito Redutase/química , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Linhagem Celular Tumoral , Chaperonina 10/química , Chaperonina 60/química , Citosol/metabolismo , Dano ao DNA , Humanos , Mitocôndrias/metabolismo , Modelos Biológicos , Mutação , Ligação Proteica , Espécies Reativas de Oxigênio
18.
Am J Pathol ; 169(2): 515-27, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16877353

RESUMO

Mutations in the gene encoding matrilin-3 (MATN3), a noncollagenous extracellular matrix protein, have been reported in a variety of skeletal diseases, including multiple epiphyseal dysplasia, which is characterized by irregular ossification of the epiphyses and early-onset osteoarthritis, spondylo-epimetaphyseal dysplasia, and idiopathic hand osteoarthritis. To assess the role of matrilin-3 in the pathogenesis of these diseases, we generated Matn3 functional knockout mice using embryonic stem cell technology. In the embryonic growth plate of the developing long bones, Matn3 null chondrocytes prematurely became prehypertrophic and hypertrophic, forming an expanded zone of hypertrophy. This expansion was attenuated during the perinatal period, and Matn3 homozygous null mice were viable and showed no gross skeletal malformations at birth. However, by 18 weeks of age, Matn3 null mice had a significantly higher total body bone mineral density than Matn1 null mice or wild-type littermates. Aged Matn3 null mice were much more predisposed to develop severe osteoarthritis than their wild-type littermates. Here, we show that matrilin-3 plays a role in modulating chondrocyte differentiation during embryonic development, in controlling bone mineral density in adulthood, and in preventing osteoarthritis during aging. The lack of Matn3 does not lead to postnatal chondrodysplasia but accounts for higher incidence of osteoarthritis.


Assuntos
Densidade Óssea/fisiologia , Condrócitos/patologia , Proteínas da Matriz Extracelular/deficiência , Osteoartrite/patologia , Envelhecimento , Animais , Animais Recém-Nascidos , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário , Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica , Marcação de Genes , Genótipo , Lâmina de Crescimento/patologia , Lâmina de Crescimento/ultraestrutura , Hipertrofia/patologia , Articulação do Joelho/diagnóstico por imagem , Articulação do Joelho/patologia , Proteínas Matrilinas , Camundongos , Camundongos Knockout , Anormalidades Musculoesqueléticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Radiografia , Tíbia/patologia , Tíbia/ultraestrutura
19.
Mol Cell Biol ; 26(1): 238-49, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16354695

RESUMO

Procollagen C proteinases (pCPs) cleave type I to III procollagen C propeptides as a necessary step in assembling the major fibrous components of vertebrate extracellular matrix. The protein PCOLCE1 (procollagen C proteinase enhancer 1) is not a proteinase but can enhance the activity of pCPs approximately 10-fold in vitro and has reported roles in inhibiting other proteinases and in growth control. Here we have generated mice with null alleles of the PCOLCE1 gene, Pcolce, to ascertain in vivo roles. Although Pcolce-/- mice are viable and fertile, Pcolce-/- male, but not female, long bones are more massive and have altered geometries that increase resistance to loading, compared to wild type. Mechanical testing indicated inferior material properties of Pcolce-/- male long bone, apparently compensated for by the adaptive changes in bone geometry. Male and female Pcolce-/- vertebrae both appeared to compensate for inferior material properties with thickened and more numerous trabeculae and had a uniquely altered morphology in deposited mineral. Ultrastructurally, Pcolce-/- mice had profoundly abnormal collagen fibrils in both mineralized and nonmineralized tissues. In Pcolce-/- tendon, 100% of collagen fibrils had deranged morphologies, indicating marked functional effects in this tissue. Thus, PCOLCE1 is an important determinant of bone mechanical properties and geometry and of collagen fibril morphology in mammals, and the human PCOLCE1 gene is identified as a candidate for phenotypes with defects in such attributes in humans.


Assuntos
Osso e Ossos/anatomia & histologia , Colágeno Tipo V/metabolismo , Tecido Conjuntivo/ultraestrutura , Glicoproteínas/genética , Pró-Colágeno/metabolismo , Alelos , Animais , Biomarcadores/análise , Fenômenos Biomecânicos , Colágeno Tipo V/ultraestrutura , Tecido Conjuntivo/química , Tecido Conjuntivo/crescimento & desenvolvimento , Proteínas da Matriz Extracelular , Feminino , Marcação de Genes , Glicoproteínas/análise , Glicoproteínas/fisiologia , Masculino , Camundongos , Camundongos Mutantes , Mutação , Peptídeos/metabolismo , Fenótipo
20.
Mol Cell Biol ; 25(18): 8299-310, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16135817

RESUMO

Pseudoxanthoma elasticum (PXE), characterized by connective tissue mineralization of the skin, eyes, and cardiovascular system, is caused by mutations in the ABCC6 gene. ABCC6 encodes multidrug resistance-associated protein 6 (MRP6), which is expressed primarily in the liver and kidneys. Mechanisms producing ectopic mineralization as a result of these mutations remain unclear. To elucidate this complex disease, a transgenic mouse was generated by targeted ablation of the mouse Abcc6 gene. Abcc6 null mice were negative for Mrp6 expression in the liver, and complete necropsies revealed profound mineralization of several tissues, including skin, arterial blood vessels, and retina, while heterozygous animals were indistinguishable from the wild-type mice. Particularly striking was the mineralization of vibrissae, as confirmed by von Kossa and alizarin red stains. Electron microscopy revealed mineralization affecting both elastic structures and collagen fibers. Mineralization of vibrissae was noted as early as 5 weeks of age and was progressive with age in Abcc6(-/-) mice but was not observed in Abcc6(+/-) or Abcc6(+/+) mice up to 2 years of age. A total body computerized tomography scan of Abcc6(-/-) mice revealed mineralization in skin and subcutaneous tissue as well as in the kidneys. These data demonstrate aberrant mineralization of soft tissues in PXE-affected organs, and, consequently, these mice recapitulate features of this complex disease.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Calcinose/patologia , Tecido Conjuntivo/patologia , Modelos Animais de Doenças , Camundongos Transgênicos , Pseudoxantoma Elástico/patologia , Animais , Biomarcadores , Calcinose/etiologia , Tecido Conjuntivo/ultraestrutura , Colágenos Fibrilares/ultraestrutura , Inativação Gênica , Marcação de Genes , Camundongos , Minerais/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos , Pseudoxantoma Elástico/etiologia , Pseudoxantoma Elástico/genética , Pele/diagnóstico por imagem , Pele/patologia , Tomografia Computadorizada por Raios X , Vibrissas/patologia , Vibrissas/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA