Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 79(3): 144, 2022 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-35188596

RESUMO

In the cornea, the epithelial basement membrane (EBM) and corneal endothelial Descemet's basement membrane (DBM) critically regulate the localization, availability and, therefore, the functions of transforming growth factor (TGF)ß1, TGFß2, and platelet-derived growth factors (PDGF) that modulate myofibroblast development. Defective regeneration of the EBM, and notably diminished perlecan incorporation, occurs via several mechanisms and results in excessive and prolonged penetration of pro-fibrotic growth factors into the stroma. These growth factors drive mature myofibroblast development from both corneal fibroblasts and bone marrow-derived fibrocytes, and then the persistence of these myofibroblasts and the disordered collagens and other matrix materials they produce to generate stromal scarring fibrosis. Corneal stromal fibrosis often resolves completely if the inciting factor is removed and the BM regenerates. Similar defects in BM regeneration are likely associated with the development of fibrosis in other organs where perlecan has a critical role in the modulation of signaling by TGFß1 and TGFß2. Other BM components, such as collagen type IV and collagen type XIII, are also critical regulators of TGF beta (and other growth factors) in the cornea and other organs. After injury, BM components are dynamically secreted and assembled through the cooperation of neighboring cells-for example, the epithelial cells and keratocytes for the corneal EBM and corneal endothelial cells and keratocytes for the corneal DBM. One of the most critical functions of these reassembled BMs in all organs is to modulate the pro-fibrotic effects of TGFßs, PDGFs and other growth factors between tissues that comprise the organ.


Assuntos
Membrana Basal/patologia , Doenças da Córnea/patologia , Fibrose/patologia , Proteoglicanas de Heparan Sulfato/deficiência , Fator de Crescimento Transformador beta/metabolismo , Animais , Membrana Basal/metabolismo , Doenças da Córnea/genética , Doenças da Córnea/metabolismo , Fibrose/genética , Fibrose/metabolismo , Humanos , Fator de Crescimento Transformador beta/genética
2.
Mol Cell Proteomics ; 19(7): 1220-1235, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32381549

RESUMO

Perlecan is a critical proteoglycan found in the extracellular matrix (ECM) of cartilage. In healthy cartilage, perlecan regulates cartilage biomechanics and we previously demonstrated perlecan deficiency leads to reduced cellular and ECM stiffness in vivo This change in mechanics may lead to the early onset osteoarthritis seen in disorders resulting from perlecan knockdown such as Schwartz-Jampel syndrome (SJS). To identify how perlecan knockdown affects the material properties of developing cartilage, we used imaging and liquid chromatography-tandem mass spectrometry (LC-MS/MS) to study the ECM in a murine model of SJS, Hspg2C1532Y-Neo Perlecan knockdown led to defective pericellular matrix formation, whereas the abundance of bulk ECM proteins, including many collagens, increased. Post-translational modifications and ultrastructure of collagens were not significantly different; however, LC-MS/MS analysis showed more protein was secreted by Hspg2C1532Y-Neo cartilage in vitro, suggesting that the incorporation of newly synthesized ECM was impaired. In addition, glycosaminoglycan deposition was atypical, which may explain the previously observed decrease in mechanics. Overall, these findings provide insight into the influence of perlecan on functional cartilage assembly and the progression of osteoarthritis in SJS.


Assuntos
Cartilagem/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Osteocondrodisplasias/metabolismo , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Cartilagem/crescimento & desenvolvimento , Cartilagem/ultraestrutura , Moléculas de Adesão Celular/metabolismo , Condrócitos/citologia , Condrócitos/metabolismo , Cromatografia Líquida , Colágeno Tipo X/genética , Colágeno Tipo X/metabolismo , Modelos Animais de Doenças , Matriz Extracelular/patologia , Ontologia Genética , Glicosaminoglicanos/metabolismo , Proteoglicanas de Heparan Sulfato/deficiência , Proteoglicanas de Heparan Sulfato/genética , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Osteoartrite/genética , Osteoartrite/metabolismo , Osteoartrite/patologia , Osteocondrodisplasias/genética , Espectrometria de Massas em Tandem
3.
Int J Mol Sci ; 23(1)2021 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-35008739

RESUMO

Perlecan (HSPG2), a basement membrane-type heparan sulfate proteoglycan, has been implicated in the development of aortic tissue. However, its role in the development and maintenance of the aortic wall remains unknown. Perlecan-deficient mice (Hspg2-/--Tg: Perl KO) have been found to show a high frequency (15-35%) of aortic dissection (AD). Herein, an analysis of the aortic wall of Perl KO mice revealed that perlecan deficiency caused thinner and partially torn elastic lamina. Compared to the control aortic tissue, perlecan-deficient aortic tissue showed a significant decrease in desmosine content and an increase in soluble tropoelastin levels, implying the presence of immature elastic fibers in Perl KO mice. Furthermore, the reduced expression of the smooth muscle cell contractile proteins actin and myosin in perlecan-deficient aortic tissue may explain the risk of AD. This study showed that a deficiency in perlecan, which is localized along the elastic lamina and at the interface between elastin and fibrillin-1, increased the risk of AD, largely due to the immaturity of extracellular matrix in the aortic tissue. Overall, we proposed a new model of AD that considers the deficiency of extracellular molecule perlecan as a risk factor.


Assuntos
Dissecção Aórtica/metabolismo , Dissecção Aórtica/patologia , Proteoglicanas de Heparan Sulfato/deficiência , Animais , Aorta/metabolismo , Aorta/patologia , Aorta/ultraestrutura , Biomarcadores/metabolismo , Elasticidade , Elastina/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Fibrilina-1/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Metaloproteinases da Matriz/metabolismo , Camundongos Transgênicos , Contração Miocárdica , Miócitos de Músculo Liso/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Risco
4.
J Cell Physiol ; 233(4): 3176-3194, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28833096

RESUMO

We investigated the role of glycosaminoglycans (GAGs) in the regulation of endothelial nitric oxide synthase (eNOS) activity in wild-type CHO-K1 cells and in xylosyltransferase-deficient CHO-745 cells. GAGs inhibit the integrin/FAK/PI3K/AKT signaling pathway in CHO-K1 cells, decreasing the phosphorylation of eNOS at Ser1177. Furthermore, in CHO-K1 cells, eNOS and PKCα are localized at sphingolipid- and cholesterol-rich domains in the plasma membrane called caveolae. At caveolae, PKCα activation stimulates the phosphorylation of eNOS on Thr495, resulting in further inhibition of NO production in these cells. In our data, CHO-745 cells generate approximately 12-fold more NO than CHO-K1 cells. Increased NO production in CHO-745 cells promotes higher rates of protein S-nitrosylation and protein tyrosine nitration. Regarding reactive oxygen species (ROS) production, CHO-745 cells show lower basal levels of superoxide (O2- ) than CHO-K1 cells. In addition, CHO-745 cells express higher levels of GPx, Trx1, and catalase than CHO-K1 cells, suggesting that CHO-745 cells are in a constitutive nitrosative/oxidative stress condition. Accordingly, we showed that CHO-745 cells are more sensitive to oxidant-induced cell death than CHO-K1 cells. The high concentration of NO and reactive oxygen species generated by CHO-745 cells can induce simultaneous mitochondrial biogenesis and antioxidant gene expression. These observations led us to propose that GAGs are part of a regulatory mechanism that participates in eNOS activation and consequently regulates nitrosative/oxidative stress in CHO cells.


Assuntos
Proteoglicanas de Heparan Sulfato/deficiência , Espaço Intracelular/metabolismo , Óxido Nítrico/biossíntese , Regulação para Cima , Animais , Células CHO , Cricetinae , Cricetulus , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Oligopeptídeos/metabolismo , Biogênese de Organelas , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Frações Subcelulares/metabolismo
5.
Nephrol Dial Transplant ; 33(1): 26-33, 2018 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-28992095

RESUMO

BACKGROUND: Heparan sulphate proteoglycan (HSPG) is present in the glomerular basement membrane (GBM) and is thought to play a major role in the glomerular charge barrier. Reductions and structural alterations of HSPG are observed in different types of kidney diseases accompanied by proteinuria. However, their causal relations remain unknown. METHODS: We generated podocyte-specific exostosin-like 3 gene (Extl3) knockout mice (Extl3KO) using a Cre-loxP recombination approach. A reduction of HSPG was expected in the GBM of these mice, because EXTL3 is involved in its synthesis. Mice were separated into three groups, according to the loads on the glomeruli: a high-protein diet group, a high-protein and high-sodium diet group and a hyperglycaemic group induced by streptozotocin treatment in addition to maintenance on a high-protein and high-sodium diet. The urinary albumin:creatinine ratio was measured at 7, 11, 15 and 19 weeks of age. Renal histology was also investigated. RESULTS: Podocyte-specific expression of Cre recombinase was detected by immunohistochemistry. Moreover, immunofluorescent staining demonstrated a significant reduction of HSPG in the GBM. Electron microscopy showed irregularities in the GBM and effacement of the foot processes in Extl3KO. The values of the urinary albumin:creatinine ratio were within the range of microalbuminuria in all groups and did not significantly differ between the control mice and Extl3KO. CONCLUSIONS: The reduction of HSPG in the GBM did not augment urinary albumin excretion. HSPG's anionic charge appears to contribute little to the glomerular charge barrier.


Assuntos
Albuminas/metabolismo , Membrana Basal Glomerular/metabolismo , Proteoglicanas de Heparan Sulfato/deficiência , Glomérulos Renais/metabolismo , N-Acetilglucosaminiltransferases/fisiologia , Podócitos/metabolismo , Urinálise , Animais , Masculino , Camundongos , Camundongos Knockout
6.
Dev Biol ; 418(2): 242-7, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27578148

RESUMO

The pericellular matrix (PCM) is a component of the extracellular matrix that is found immediately surrounding individual chondrocytes in developing and adult cartilage, and is rich in the proteoglycan perlecan. Mutations in perlecan are the basis of several developmental disorders, which are thought to arise from disruptions in the mechanical stability of the PCM. We tested the hypothesis that defects in PCM organization will reduce the stiffness of chondrocytes in developing cartilage by combining a murine model of Schwartz-Jampel syndrome, in which perlecan is knocked down, with our novel atomic force microscopy technique that can measure the stiffness of living cells and surrounding matrix in embryonic and postnatal tissues in situ. Perlecan knockdown altered matrix organization and significantly decreased the stiffness of both chondrocytes and interstitial matrix as a function of age and genotype. Our results demonstrate that the knockdown of a spatially restricted matrix molecule can have a profound influence on cell and tissue stiffness, implicating a role for outside-in mechanical signals from the PCM in regulating the intracellular mechanisms required for the overall development of cartilage.


Assuntos
Cartilagem/fisiopatologia , Proteínas da Matriz Extracelular/deficiência , Proteoglicanas de Heparan Sulfato/deficiência , Animais , Fenômenos Biomecânicos , Cartilagem/crescimento & desenvolvimento , Cartilagem/patologia , Condrócitos/patologia , Condrócitos/fisiologia , Modelos Animais de Doenças , Matriz Extracelular/patologia , Matriz Extracelular/fisiologia , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/fisiologia , Feminino , Técnicas de Silenciamento de Genes , Proteoglicanas de Heparan Sulfato/genética , Proteoglicanas de Heparan Sulfato/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Microscopia de Força Atômica , Osteocondrodisplasias/genética , Osteocondrodisplasias/patologia , Osteocondrodisplasias/fisiopatologia , Gravidez
7.
Calcif Tissue Int ; 95(1): 29-38, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24798737

RESUMO

Perlecan/HSPG2 (Pln) is a large heparan sulfate proteoglycan abundant in the extracellular matrix of cartilage and the lacunocanalicular space of adult bones. Although Pln function during cartilage development is critical, evidenced by deficiency disorders including Schwartz-Jampel Syndrome and dyssegmental dysplasia Silverman-Handmaker type, little is known about its function in development of bone shape and quality. The purpose of this study was to understand the contribution of Pln to bone geometric and mechanical properties. We used hypomorph mutant mice that secrete negligible amount of Pln into skeletal tissues and analyzed their adult bone properties using micro-computed tomography and three-point-bending tests. Bone shortening and widening in Pln mutants was observed and could be attributed to loss of growth plate organization and accelerated osteogenesis that was reflected by elevated cortical thickness at older ages. This effect was more pronounced in Pln mutant females, indicating a sex-specific effect of Pln deficiency on bone geometry. Additionally, mutant females, and to a lesser extent mutant males, increased their elastic modulus and bone mineral densities to counteract changes in bone shape, but at the expense of increased brittleness. In summary, Pln deficiency alters cartilage matrix patterning and, as we now show, coordinately influences bone formation and calcification.


Assuntos
Desenvolvimento Ósseo/fisiologia , Proteoglicanas de Heparan Sulfato/deficiência , Osteogênese/fisiologia , Envelhecimento , Animais , Osso e Ossos , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microscopia Confocal , Microtomografia por Raio-X
8.
Histochem Cell Biol ; 139(1): 1-11, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23104139

RESUMO

The aim of this study was to examine the comparative localisations of fibrillin-1 and perlecan in the foetal human, wild-type C57BL/6 and HS-deficient hspg2Δ³â»/Δ³â» exon 3 null mouse intervertebral disc (IVD) using fluorescent laser scanning confocal microscopy. Fibrillin-1 fibrils were prominent components of the outer posterior and anterior annulus fibrosus (AF) of the foetal human IVD. Finer fibrillin-1 fibrils were evident in the inner AF where they displayed an arcade-type arrangement in the developing lamellae. Relatively short but distinct fibrillin-1 fibrils were evident in the central region of the IVD and presumptive cartilaginous endplate and defined the margins of the nuclear sheath in the developing nucleus pulposus (NP). Fibrillin-1 was also demonstrated in the AF of C57BL/6 wild-type mice but to a far lesser extent in the HS-deficient hspg2Δ³â»/Δ³â» exon 3 null mouse. This suggested that the HS chains of perlecan may have contributed to fibrillin-1 assembly or its deposition in the IVD. The cell-matrix interconnections provided by the fibrillin fibrils visualised in this study may facilitate communication between disc cells and their local biomechanical microenvironment in mechanosensory processes which regulate tissue homeostasis. The ability of fibrillin-1 to sequester TGF-ß a well-known anabolic growth factor in the IVD also suggests potential roles in disc development and/or remodelling.


Assuntos
Proteoglicanas de Heparan Sulfato/deficiência , Imuno-Histoquímica , Disco Intervertebral/metabolismo , Proteínas dos Microfilamentos/metabolismo , Mutação , Animais , Éxons , Fibrilina-1 , Fibrilinas , Idade Gestacional , Proteoglicanas de Heparan Sulfato/genética , Humanos , Disco Intervertebral/embriologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Microscopia de Fluorescência
9.
Am J Pathol ; 180(5): 2040-55, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22449950

RESUMO

Congenital peripheral nerve hyperexcitability (PNH) is usually associated with impaired function of voltage-gated K(+) channels (VGKCs) in neuromyotonia and demyelination in peripheral neuropathies. Schwartz-Jampel syndrome (SJS) is a form of PNH that is due to hypomorphic mutations of perlecan, the major proteoglycan of basement membranes. Schwann cell basement membrane and its cell receptors are critical for the myelination and organization of the nodes of Ranvier. We therefore studied a mouse model of SJS to determine whether a role for perlecan in these functions could account for PNH when perlecan is lacking. We revealed a role for perlecan in the longitudinal elongation and organization of myelinating Schwann cells because perlecan-deficient mice had shorter internodes, more numerous Schmidt-Lanterman incisures, and increased amounts of internodal fast VGKCs. Perlecan-deficient mice did not display demyelination events along the nerve trunk but developed dysmyelination of the preterminal segment associated with denervation processes at the neuromuscular junction. Investigating the excitability properties of the peripheral nerve suggested a persistent axonal depolarization during nerve firing in vitro, most likely due to defective K(+) homeostasis, and excluded the nerve trunk as the original site for PNH. Altogether, our data shed light on perlecan function by revealing critical roles in Schwann cell physiology and suggest that PNH in SJS originates distally from synergistic actions of peripheral nerve and neuromuscular junction changes.


Assuntos
Axônios/fisiologia , Proteoglicanas de Heparan Sulfato/fisiologia , Osteocondrodisplasias/patologia , Células de Schwann/fisiologia , Potenciais de Ação/fisiologia , Envelhecimento/fisiologia , Animais , Membrana Basal/metabolismo , Doenças Desmielinizantes/etiologia , Modelos Animais de Doenças , Estimulação Elétrica/métodos , Proteoglicanas de Heparan Sulfato/deficiência , Proteoglicanas de Heparan Sulfato/genética , Canal de Potássio Kv1.1/biossíntese , Camundongos , Camundongos Mutantes , Microscopia Eletrônica , Mutação , Bainha de Mielina/fisiologia , Bainha de Mielina/ultraestrutura , Junção Neuromuscular/fisiopatologia , Osteocondrodisplasias/complicações , Osteocondrodisplasias/fisiopatologia , Nós Neurofibrosos/metabolismo , Nós Neurofibrosos/ultraestrutura , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Células de Schwann/metabolismo , Nervo Isquiático/fisiopatologia , Nervo Isquiático/ultraestrutura
10.
J Neurosci ; 31(5): 1644-51, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21289173

RESUMO

Alzheimer's disease (AD) is a progressive and irreversible neurodegenerative disorder in which the aggregation and deposition of amyloid-ß (Aß) peptides in the brain are central to its pathogenesis. In healthy brains, Aß is effectively metabolized with little accumulation. Cellular uptake and subsequent degradation of Aß is one of the major pathways for its clearance in the brain. Increasing evidence has demonstrated significant roles for the low-density lipoprotein receptor-related protein 1 (LRP1) in the metabolism of Aß in neurons, glia cells, and along the brain vasculatures. Heparan sulfate proteoglycan (HSPG) has also been implicated in several pathogenic features of AD, including its colocalization with amyloid plaques. Here, we demonstrate that HSPG and LRP1 cooperatively mediate cellular Aß uptake. Fluorescence-activated cell sorter and confocal microscopy revealed that knockdown of LRP1 suppresses Aß uptake, whereas overexpression of LRP1 enhances this process in neuronal cells. Heparin, which antagonizes HSPG, significantly inhibited cellular Aß uptake. Importantly, treatment with heparin or heparinase blocked LRP1-mediated cellular uptake of Aß. We further showed that HSPG is more important for the binding of Aß to the cell surface than LRP1. The critical roles of HSPG in cellular Aß binding and uptake were confirmed in Chinese hamster ovary cells genetically deficient in HSPG. We also showed that heparin and a neutralizing antibody to LRP1 suppressed Aß uptake in primary neurons. Our findings demonstrate that LRP1 and HSPG function in a cooperative manner to mediate cellular Aß uptake and define a major pathway through which Aß gains entry to neuronal cells.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Fibroblastos/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Transporte Biológico , Western Blotting , Células CHO , Linhagem Celular , Células Cultivadas , Cricetinae , Cricetulus , Embrião de Mamíferos , Endocitose/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Feminino , Fibroblastos/efeitos dos fármacos , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Proteoglicanas de Heparan Sulfato/antagonistas & inibidores , Proteoglicanas de Heparan Sulfato/deficiência , Proteoglicanas de Heparan Sulfato/genética , Heparina/farmacologia , Hipotálamo/citologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Microscopia Confocal , Neurônios/efeitos dos fármacos , Gravidez , RNA Interferente Pequeno , Receptores de LDL/genética , Transfecção , Proteínas Supressoras de Tumor/genética , Regulação para Cima/efeitos dos fármacos
11.
J Biol Chem ; 285(3): 1861-9, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19897482

RESUMO

In semen, proteolytic peptide fragments from prostatic acid phosphatase can form amyloid fibrils termed SEVI (semen-derived enhancer of viral infection). These fibrils greatly enhance human immunodeficiency virus (HIV) infectivity by increasing the attachment of virions to target cells. Therefore, SEVI may have a significant impact on whether HIV is successfully transmitted during sexual contact. Here, we demonstrate that surfen, a small molecule heparan sulfate proteoglycan antagonist, inhibits both SEVI- and semen-mediated enhancement of HIV type 1 infection. Surfen interferes with the binding of SEVI to both target cells and HIV type 1 virions but does not deaggregate SEVI fibrils. Because SEVI can increase HIV infectivity by several orders of magnitude, supplementing current HIV microbicide candidates with SEVI inhibitors, such as surfen, might greatly increase their potency.


Assuntos
Aminoquinolinas/farmacologia , Amiloide/antagonistas & inibidores , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Sêmen/efeitos dos fármacos , Sêmen/metabolismo , Ureia/análogos & derivados , Aminoquinolinas/uso terapêutico , Amiloide/metabolismo , Animais , Fármacos Anti-HIV/farmacologia , Fármacos Anti-HIV/uso terapêutico , Linhagem Celular , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Infecções por HIV/metabolismo , HIV-1/metabolismo , HIV-1/fisiologia , Proteoglicanas de Heparan Sulfato/deficiência , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Camundongos , Ratos , Sêmen/virologia , Ureia/farmacologia , Ureia/uso terapêutico , Vírion/efeitos dos fármacos , Vírion/metabolismo , Internalização do Vírus/efeitos dos fármacos
12.
Hum Mol Genet ; 17(20): 3166-79, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18647752

RESUMO

Schwartz-Jampel syndrome (SJS) is a recessive neuromyotonia with chondrodysplasia. It results from hypomorphic mutations of the gene encoding perlecan, leading to a decrease in the levels of this heparan sulphate proteoglycan in basement membranes (BMs). It has been suggested that SJS neuromyotonia may result from endplate acetylcholinesterase (AChE) deficiency, but this hypothesis has never been investigated in vivo due to the lack of an animal model for neuromyotonia. We used homologous recombination to generate a knock-in mouse strain with one missense substitution, corresponding to a human familial SJS mutation (p.C1532Y), in the perlecan gene. We derived two lines, one with the p.C1532Y substitution alone and one with p.C1532Y and the selectable marker Neo, to down-regulate perlecan gene activity and to test for a dosage effect of perlecan in mammals. These two lines mimicked SJS neuromyotonia with spontaneous activity on electromyogramm (EMG). An inverse correlation between disease severity and perlecan secretion in the BMs was observed at the macroscopic and microscopic levels, consistent with a dosage effect. Endplate AChE levels were low in both lines, due to synaptic perlecan deficiency rather than major myofibre or neuromuscular junction disorganization. Studies of muscle contractile properties showed muscle fatigability at low frequencies of nerve stimulation and suggested that partial endplate AChE deficiency might contribute to SJS muscle stiffness by potentiating muscle force. However, physiological endplate AChE deficiency was not associated with spontaneous activity at rest on EMG in the diaphragm, suggesting that additional changes are required to generate such activity characteristic of SJS.


Assuntos
Acetilcolinesterase/deficiência , Acetilcolinesterase/genética , Síndrome de Isaacs/enzimologia , Síndrome de Isaacs/genética , Placa Motora/enzimologia , Osteocondrodisplasias/enzimologia , Osteocondrodisplasias/genética , Alelos , Animais , Modelos Animais de Doenças , Eletromiografia , Feminino , Dosagem de Genes , Proteoglicanas de Heparan Sulfato/deficiência , Proteoglicanas de Heparan Sulfato/genética , Humanos , Síndrome de Isaacs/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Placa Motora/fisiopatologia , Contração Muscular/genética , Contração Muscular/fisiologia , Mutação de Sentido Incorreto , Osteocondrodisplasias/fisiopatologia , Fenótipo
13.
Biomolecules ; 10(2)2020 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-32013135

RESUMO

Perlecan/Hspg2, a large monomeric heparan sulfate proteoglycan, is found in the basement membrane and extracellular matrix, where it acts as a matrix scaffold, growth factor depot, and tissue barrier. Perlecan deficiency leads to skeletal dysplasia in Schwartz-Jampel Syndrome (SJS) and is a risk factor for osteoporosis. In the SJS-mimicking murine model (Hypo), inferior cortical bone quality and impaired mechanotransduction in osteocytes were reported. This study focused on trabecular bone, where perlecan deficiency was hypothesized to result in structural deficit and altered response to disuse and re-loading. We compared the Hypo versus WT trabecular bone in both axial and appendicular skeletons of 8-38-week-old male mice, and observed severe trabecular deficit in Hypo mice, approximately 50% reduction of Tb.BV/TV regardless of skeletal site and animal age. Defects in endochondral ossification (e.g., accelerated mineralization), increases in osteoclast activity, and altered differentiation of bone progenitor cells in marrow contributed to the Hypo phenotype. The Hypo trabecular bone deteriorated further under three-week hindlimb suspension as did the WT. Re-ambulation partially recovered the lost trabecular bone in Hypo, but not in WT mice. The novel finding that low-impact loading could counter detrimental disuse effects in the perlecan-deficient skeleton suggests a strategy to maintain skeletal health in SJS patients.


Assuntos
Osso Esponjoso/patologia , Proteoglicanas de Heparan Sulfato/deficiência , Proteoglicanas de Heparan Sulfato/genética , Osteócitos/citologia , Animais , Fêmur/patologia , Células-Tronco Hematopoéticas/citologia , Proteoglicanas de Heparan Sulfato/fisiologia , Cifose , Masculino , Mecanotransdução Celular , Metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos/citologia , Osteogênese , Fenótipo , Fatores de Risco , Estresse Mecânico , Caminhada , Microtomografia por Raio-X
14.
Protein Eng Des Sel ; 32(2): 95-102, 2019 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-31769491

RESUMO

Islet amyloid is a pathologic feature of type 2 diabetes (T2D) that is associated with ß-cell loss and dysfunction. These amyloid deposits form via aggregation of the ß-cell secretory product islet amyloid polypeptide (IAPP) and contain other molecules including the heparan sulfate proteoglycan perlecan. Perlecan has been shown to bind amyloidogenic human IAPP (hIAPP) via its heparan sulfate glycosaminoglycan (HS GAG) chains and to enhance hIAPP aggregation in vitro. We postulated that reducing the HS GAG content of perlecan would also decrease islet amyloid deposition in vivo. hIAPP transgenic mice were crossed with Hspg2Δ3/Δ3 mice harboring a perlecan mutation that prevents HS GAG attachment (hIAPP;Hspg2Δ3/Δ3), and male offspring from this cross were fed a high fat diet for 12 months to induce islet amyloid deposition. At the end of the study body weight, islet amyloid area, ß-cell area, glucose tolerance and insulin secretion were analyzed. hIAPP;Hspg2Δ3/Δ3 mice exhibited significantly less islet amyloid deposition and greater ß-cell area compared to hIAPP mice expressing wild type perlecan. hIAPP;Hspg2Δ3/Δ3 mice also gained significantly less weight than other genotypes. When adjusted for differences in body weight using multiple linear regression modeling, we found no differences in islet amyloid deposition or ß-cell area between hIAPP transgenic and hIAPP;Hspg2Δ3/Δ3 mice. We conclude that loss of perlecan exon 3 reduces islet amyloid deposition in vivo through indirect effects on body weight and possibly also through direct effects on hIAPP aggregation. Both of these mechanisms may promote maintenance of glucose homeostasis in the setting of T2D.


Assuntos
Peso Corporal , Proteoglicanas de Heparan Sulfato/deficiência , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Animais , Contagem de Células , Humanos , Camundongos , Camundongos Transgênicos
15.
J Cell Biol ; 218(10): 3506-3525, 2019 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-31541017

RESUMO

Ischemic stroke causes blood-brain barrier (BBB) breakdown due to significant damage to the integrity of BBB components. Recent studies have highlighted the importance of pericytes in the repair process of BBB functions triggered by PDGFRß up-regulation. Here, we show that perlecan, a major heparan sulfate proteoglycan of basement membranes, aids in BBB maintenance and repair through pericyte interactions. Using a transient middle cerebral artery occlusion model, we found larger infarct volumes and more BBB leakage in conditional perlecan (Hspg2)-deficient (Hspg2 - / - -TG) mice than in control mice. Control mice showed increased numbers of pericytes in the ischemic lesion, whereas Hspg2 - / - -TG mice did not. At the mechanistic level, pericytes attached to recombinant perlecan C-terminal domain V (perlecan DV, endorepellin). Perlecan DV enhanced the PDGF-BB-induced phosphorylation of PDGFRß, SHP-2, and FAK partially through integrin α5ß1 and promoted pericyte migration. Perlecan therefore appears to regulate pericyte recruitment through the cooperative functioning of PDGFRß and integrin α5ß1 to support BBB maintenance and repair following ischemic stroke.


Assuntos
Barreira Hematoencefálica/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Pericitos/metabolismo , Animais , Barreira Hematoencefálica/patologia , Modelos Animais de Doenças , Proteoglicanas de Heparan Sulfato/administração & dosagem , Proteoglicanas de Heparan Sulfato/deficiência , Infarto da Artéria Cerebral Média/patologia , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
16.
BMC Dev Biol ; 7: 29, 2007 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-17411441

RESUMO

BACKGROUND: Perlecan is a proteoglycan expressed in the basal lamina of the neuroepithelium during development. Perlecan absence does not impair basal lamina assembly, although in the 55% of the mutants early disruptions of this lamina conducts to exencephaly, impairing brain development. The rest of perlecan-null brains complete its prenatal development, maintain basal lamina continuity interrupted by some isolated ectopias, and are microcephalic. Microcephaly consists of thinner cerebral walls and underdeveloped ganglionic eminences. We have studied the mechanisms that generate brain atrophy in telencephalic areas where basal lamina is intact. RESULTS: Brain atrophy in the absence of perlecan started in the ventral forebrain and extended to lateral and dorsal parts of the cortex in the following stages. First, the subpallial forebrain developed poorly in early perlecan-null embryos, because of a reduced cell proliferation: the number of cells in mitosis decreased since the early stages of development. This reduction resulted in a decreased tangential migration of interneurons to the cerebral cortex. Concomitant with the early hypoplasia observed in the medial ganglionic eminences, Sonic Hedgehog signal decreased in the perlecan-null floor plate basal lamina at E12.5. Second, neurogenesis in the pallial neuroepithelium was affected in perlecan deficient embryos. We found reductions of nearly 50% in the number of cells exiting the cell cycle at E12-E13. The labeling index, which was normal at this age, significantly decreased with advancing corticogenesis. Moreover, nestin+ or PCNA+ progenitors increased since E14.5, reaching up to about 150% of the proportion of PCNA+ cells in the wild-type at E17.5. Thus, labeling index reduction together with increased progenitor population, suggests that atrophy is the result of altered cell cycle progression in the cortical progenitors. Accordingly, less neurons populated the cortical plate and subplate of perlecan-null neocortex, as seen with the neuronal markers beta-tubulin and Tbr1. CONCLUSION: As a component of the basal lamina, perlecan both maintains this structure and controls the response of the neuroepithelium to growth factors. Less mitotic cells in the early medial ganglionic eminences, and impaired cell cycle progression in the late neocortex, suggests insufficient recruitment and signaling by neurogenic morphogens, such as SHH or FGF2.


Assuntos
Proteoglicanas de Heparan Sulfato/genética , Organogênese , Telencéfalo/embriologia , Animais , Apoptose , Membrana Basal/citologia , Membrana Basal/embriologia , Membrana Basal/metabolismo , Contagem de Células , Proliferação de Células , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Maturidade dos Órgãos Fetais , Fator 2 de Crescimento de Fibroblastos/genética , Proteínas Hedgehog/genética , Proteoglicanas de Heparan Sulfato/deficiência , Imuno-Histoquímica , Interneurônios/citologia , Interneurônios/metabolismo , Masculino , Camundongos , Microcefalia/genética , Neocórtex/citologia , Neocórtex/embriologia , Neocórtex/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Gravidez , Telencéfalo/citologia , Telencéfalo/metabolismo
17.
Mech Dev ; 122(7-8): 928-38, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15925496

RESUMO

Renal branching morphogenesis, defined as growth and branching of the ureteric bud (UB), is a tightly regulated process controlled by growth factor-dependent tissue interactions. Previously, using in vitro models of branching morphogenesis, we demonstrated that BMP2 signals via its intracellular effectors, SMAD1 and SMAD4, to control UB cell proliferation and branching in a manner modulated by Glypican-3 (GPC3), a cell surface heparan sulfate proteoglycan. Here, we used loss-of-function genetic mouse models to investigate the functions of Bmp2 and Gpc3-Bmp2 interactions in vivo. Progressively greater increases in UB cell proliferation were observed in Bmp2+/-, Smad4+/-, and Bmp2+/-; Smad4+/- mice compared to Wt. This increased cell proliferation was accompanied by a significant increase in UB branching in Smad4+/- and Bmp2+/-;Smad4+/- mice compared to Wt. Reduction of Gpc3 gene dosage also increased UB cell proliferation, an effect that was enhanced in Gpc3+/-;Bmp2+/- mice to an extent greater than the sum of that observed in Gpc3+/- and Bmp2+/- mice. Reduction of both Gpc3 and Bmp2 gene dosage enhanced cell proliferation in the metanephric mesenchyme compared to Wt, an effect not observed in either Bmp2+/- or Gpc3+/- mice. Phosphorylation of SMAD1, a measure of SMAD1 activation, was progressively decreased in Gpc3+/- and Gpc3+/-;Bmp2+/- mice compared to Wt, suggesting that Gpc3 stimulates Bmp2-dependent SMAD signaling in vivo. These results demonstrate that BMP2-SMAD signaling, modulated by GPC3, inhibits renal branching morphogenesis in vivo.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Rim/embriologia , Rim/metabolismo , Transdução de Sinais , Proteína Smad1/metabolismo , Proteína Smad4/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/deficiência , Proteínas Morfogenéticas Ósseas/genética , Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento , Glipicanas , Proteoglicanas de Heparan Sulfato/deficiência , Proteoglicanas de Heparan Sulfato/genética , Rim/citologia , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Proteína Smad4/deficiência , Proteína Smad4/genética , Fator de Crescimento Transformador beta/deficiência , Fator de Crescimento Transformador beta/genética
18.
Circ Res ; 94(2): 175-83, 2004 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-14656929

RESUMO

We were interested in the elucidation of the interaction between the heparan sulfate proteoglycan, perlecan, and PTEN in the regulation of vascular smooth muscle cell (SMC) growth. We verified serum-stimulated DNA synthesis, and Akt and FAK phosphorylation were significantly reduced in SMCs overexpressing wild-type PTEN. Our previous studies showed perlecan is a potent inhibitor of serum-stimulated SMC growth. We report in the present study, compared with SMCs plated on fibronectin, serum-stimulated SMCs plated on perlecan exhibited increased PTEN activity, decreased FAK and Akt activities, and high levels of p27, consistent with SMC growth arrest. Adenoviral-mediated overexpression of constitutively active Akt reversed perlecan-induced SMC growth arrest while morpholino antisense-mediated loss of endogenous PTEN resulted in increased growth and phosphorylation of FAK and Akt of SMCs on perlecan. Immunohistochemical and Western analyses of balloon-injured rat carotid artery tissues showed a transient increase in phosphoPTEN (inactive) after injury, correlating to high rates of neointimal cell replication; phosphoPTEN was largely limited to actively replicating SMCs. Similarly, in the developing rat aorta, we found increased PTEN activity associated with increased perlecan deposition and decreased SMC replication rates. However, significantly decreased PTEN activity was detected in aortas of perlecan-deficient mouse embryos, consistent with SMC hyperplasia observed in these animals, compared with E17.5 heterozygous controls that produce abundant amounts of perlecan at this developmental time point. Our data show PTEN is a potent endogenously produced inhibitor of SMC growth and increased PTEN activity mediates perlecan-induced suppression of SMC proliferation.


Assuntos
Proteoglicanas de Heparan Sulfato/fisiologia , Músculo Liso Vascular/citologia , Proteínas Serina-Treonina Quinases , Animais , Aorta Torácica/citologia , Aorta Torácica/embriologia , Membrana Basal/fisiologia , Lesões das Artérias Carótidas/patologia , Cateterismo/efeitos adversos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura Livres de Soro , Replicação do DNA/efeitos dos fármacos , Fibronectinas/farmacologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Glicosaminoglicanos/fisiologia , Proteoglicanas de Heparan Sulfato/deficiência , Proteoglicanas de Heparan Sulfato/farmacologia , Heparitina Sulfato/fisiologia , Masculino , Camundongos , Músculo Liso Vascular/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/fisiologia
19.
Arterioscler Thromb Vasc Biol ; 25(1): 71-6, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15514208

RESUMO

OBJECTIVE: Low-molecular-weight heparin (LMWH) exerts antitumor activity in clinical trials. The K5 polysaccharide from Escherichia coli has the same structure as the heparin precursor. Chemical and enzymatic modifications of K5 polysaccharide lead to the production of biotechnological heparin-like compounds. We investigated the fibroblast growth factor-2 (FGF2) antagonist and antiangiogenic activity of a series of LMW N,O-sulfated K5 derivatives. METHODS AND RESULTS: Surface plasmon resonance analysis showed that LMW-K5 derivatives bind FGF2, thus inhibiting its interaction with heparin immobilized to a BIAcore sensor chip. Interaction of FGF2 with tyrosine-kinase receptors (FGFRs), heparan sulfate proteoglycans (HSPGs), and alpha(v)beta3 integrin is required for biological response in endothelial cells. Similar to LMWH, LMW-K5 derivatives abrogate the formation of HSPG/FGF2/FGFR ternary complexes by preventing FGF2-mediated attachment of FGFR1-overexpressing cells to HSPG-bearing cells and inhibit FGF2-mediated endothelial cell proliferation. However, LMW-K5 derivatives, but not LMWH, also inhibit FGF2/alpha(v)beta3 integrin interaction and consequent FGF2-mediated endothelial cell sprouting in vitro and angiogenesis in vivo in the chick embryo chorioallantoic membrane. CONCLUSIONS: LMW N,O-sulfated K5 derivatives affect both HSPG/FGF2/FGFR and FGF2/alpha(v)beta3 interactions and are endowed with FGF2 antagonist and antiangiogenic activity. These compounds may provide the basis for the design of novel LMW heparin-like angiostatic compounds.


Assuntos
Inibidores da Angiogênese/biossíntese , Escherichia coli/química , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Engenharia Genética/métodos , Heparina de Baixo Peso Molecular/biossíntese , Polissacarídeos Bacterianos/biossíntese , Inibidores da Angiogênese/genética , Animais , Cápsulas Bacterianas , Células CHO/química , Células CHO/metabolismo , Bovinos , Adesão Celular/fisiologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Embrião de Galinha , Membrana Corioalantoide/efeitos dos fármacos , Cricetinae , Cricetulus , Células Endoteliais/química , Células Endoteliais/metabolismo , Escherichia coli/genética , Fator 2 de Crescimento de Fibroblastos/análogos & derivados , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/análogos & derivados , Fatores de Crescimento de Fibroblastos/metabolismo , Proteoglicanas de Heparan Sulfato/análogos & derivados , Proteoglicanas de Heparan Sulfato/deficiência , Proteoglicanas de Heparan Sulfato/metabolismo , Heparina de Baixo Peso Molecular/síntese química , Heparina de Baixo Peso Molecular/genética , Integrina alfaVbeta3/metabolismo , Camundongos , Neovascularização Fisiológica/efeitos dos fármacos , Polissacarídeos Bacterianos/genética
20.
Cancer Res ; 64(14): 4699-702, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15256433

RESUMO

Perlecan, a modular proteoglycan carrying primary heparan sulfate (HS) side chains, is a major component of blood vessel basement membranes. It sequesters growth factors such as fibroblast growth factor 2 (FGF-2) and regulates the ligand-receptor interactions on the cell surface, and thus it has been implicated in the control of angiogenesis. Both stimulatory and inhibitory effects of perlecan on FGF-2 signaling have been reported. To understand the in vivo function of HS carried by perlecan, the perlecan gene heparan sulfate proteoglycan 2 (Hspg2) was mutated in mouse by gene targeting. The HS at the NH(2) terminus of perlecan was removed while the core protein remained intact. Perlecan HS-deficient (Hspg2(Delta3/Delta3)) mice survived embryonic development and were apparently healthy as adults. However, mutant mice exhibited significantly delayed wound healing, retarded FGF-2-induced tumor growth, and defective angiogenesis. In the mouse corneal angiogenesis model, FGF-2-induced neovascularization was significantly impaired in Hspg2(Delta3/Delta3) mutant mice. Our results suggest that HS in perlecan positively regulates the angiogenesis in vivo.


Assuntos
Proteoglicanas de Heparan Sulfato/deficiência , Neoplasias Experimentais/irrigação sanguínea , Neovascularização Fisiológica/fisiologia , Cicatrização/fisiologia , Animais , Membrana Basal/patologia , Vasos Sanguíneos/patologia , Divisão Celular/fisiologia , Córnea/irrigação sanguínea , Feminino , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/fisiologia , Proteoglicanas de Heparan Sulfato/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Neovascularização Patológica/patologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA