Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Osteoarthritis Cartilage ; 24(2): 335-44, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26362410

RESUMO

OBJECTIVES: The generation of transgenic mice expressing green fluorescent proteins (GFPs) has greatly aided our understanding of the development of connective tissues such as bone and cartilage. Perturbation of a biological system such as the temporomandibular joint (TMJ) within its adaptive remodeling capacity is particularly useful in analyzing cellular lineage progression. The objectives of this study were to determine: (i) if GFP reporters expressed in the TMJ indicate the different stages of cell maturation in fibrocartilage and (ii) how mechanical loading affects cellular response in different regions of the cartilage. DESIGN/METHODS: Four-week-old transgenic mice harboring combinations of fluorescent reporters (Dkk3-eGFP, Col1a1(3.6 kb)-GFPcyan, Col1a1(3.6 kb)-GFPtpz, Col2a1-GFPcyan, and Col10a1-RFPcherry) were used to analyze the expression pattern of transgenes in the mandibular condylar cartilage (MCC). To study the effect of TMJ loading, animals were subjected to forced mouth opening with custom springs exerting 50 g force for 1 h/day for 5 days. Dynamic mineralization and cellular proliferation (EdU-labeling) were assessed in loaded vs control mice. RESULTS: Dkk3 expression was seen in the superficial zone of the MCC, followed by Col1 in the cartilage zone, Col2 in the prehypertrophic zone, and Col10 in the hypertrophic zone at and below the tidemark. TMJ loading increased expression of the GFP reporters and EdU-labeling of cells in the cartilage, resulting in a thickness increase of all layers of the cartilage. In addition, mineral apposition increased resulting in Col10 expression by unmineralized cells above the tidemark. CONCLUSION: The TMJ responded to static loading by forming thicker cartilage through adaptive remodeling.


Assuntos
Condrócitos/metabolismo , Colágeno Tipo II/metabolismo , Colágeno Tipo I/metabolismo , Colágeno Tipo X/metabolismo , Fibrocartilagem/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Articulação Temporomandibular/metabolismo , Suporte de Carga , Proteínas Adaptadoras de Transdução de Sinal , Animais , Fenômenos Biomecânicos , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Linhagem da Célula , Cadeia alfa 1 do Colágeno Tipo I , Fibrocartilagem/patologia , Proteínas de Fluorescência Verde/genética , Proteínas Luminescentes/genética , Côndilo Mandibular/metabolismo , Côndilo Mandibular/patologia , Camundongos , Camundongos Transgênicos , Tamanho do Órgão , Articulação Temporomandibular/patologia , Proteína Vermelha Fluorescente
2.
Osteoarthritis Cartilage ; 23(6): 996-1006, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25680653

RESUMO

OBJECTIVE: A major challenge to understanding osteoarthritis (OA) pathology is identifying the cellular events that precede the onset of cartilage damage. The objective of this study is to determine the effect of joint destabilization on early changes to fibrocartilage in the joint. DESIGN/METHODS: The anterior cruciate ligament was transected in collagen reporter mice (Col1CFP and ColXRFP). Mineralization labels were given every 2 weeks to measure new mineralized cartilage apposition. Novel fluorescent histology of mineralized tissue was used to characterize the changes in fibrocartilage at 2 and 4 weeks post-injury. RESULTS: Changes in fibrocartilaginous structures of the joint occur as early as 2 weeks after injury and are well developed by 4 weeks. The alterations are seen in multiple entheses and in the medial surface of the femoral and tibial condyles. In the responding entheses, mineral apposition towards the ligament midsubstance results in thickening of the mineralize fibrocartilage. These changes are associated with increases in ColX-RFP, Col1-CFP reporter activity and alkaline phosphatase enzyme activity. Mineral apposition also occurs in the fibrocartilage of the non-articular regions of the medial condyles by 2 weeks and develops into osteophytes by 4 weeks post-injury. An unexpected observation is punctate expression of tartrate resistant acid phosphatase activity in unmineralized fibrochondrocytes adjacent to active appositional mineralization. DISCUSSION: These observations suggest that fibrocartilage activates prior to degradation of the articular cartilage. Thus clinical and histological imaging of fibrocartilage may be an earlier indicator of disease initiation and may indicate a more appropriate time to start preventative treatment.


Assuntos
Lesões do Ligamento Cruzado Anterior , Fibrocartilagem/fisiopatologia , Instabilidade Articular/fisiopatologia , Fosfatase Ácida/metabolismo , Animais , Calcificação Fisiológica/fisiologia , Cartilagem Articular/patologia , Condrócitos/metabolismo , Modelos Animais de Doenças , Feminino , Fêmur/patologia , Fibrocartilagem/patologia , Genes Reporter , Proteínas de Fluorescência Verde , Isoenzimas/metabolismo , Instabilidade Articular/metabolismo , Instabilidade Articular/patologia , Camundongos Transgênicos , Fosfatase Ácida Resistente a Tartarato , Tíbia/patologia
3.
Acta Physiol (Oxf) ; 208(2): 180-90, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23506283

RESUMO

AIM: Mustn1 has been implicated in myofusion as well as skeletal muscle growth and repair; however, the exact role and spatio-temporal expression of Mustn1 have yet to be fully defined. METHODS: Transgenic mice were generated with a 1512-bp sequence of the Mustn1 promoter directing the expression of GFP (Mustn1(PRO) -GFP). These mice were used to investigate the spatio-temporal expression of Mustn1(PRO) -GFP during skeletal muscle development and adult skeletal muscle repair, as well as various phases of the satellite cell lifespan (i.e. quiescence, activation, proliferation, differentiation). RESULTS: Mustn1(PRO) -GFP expression was observed within somites at embryonic day 12 and developing skeletal muscles at embryonic day 15 and 18. While uninjured adult tibialis anterior muscle displayed no detectable Mustn1(PRO) -GFP expression, cardiotoxin injury robustly elevated Mustn1(PRO) -GFP expression at 3 days post-injury with decreasing levels observed at 5 days and minimal, focal expression seen at 10 days. The expression of Mustn1(PRO) -GFP at 3 days post-injury consistently overlaid with MyoD although the strongest expression of Mustn1(PRO) -GFP was noted in newly formed myotubes that were expressing minimal levels of MyoD. By 5 days post-injury, Mustn1(PRO) -GFP overlaid in all myotubes expressing myogenin although cells were present expressing Mustn1(PRO) -GFP alone. The expression patterns of Mustn1(PRO) -GFP in regenerating muscle preceded the expression of desmin throughout the regenerative time course consistent with Mustn1 being upstream of this myogenic protein. Further, quiescent satellite cells located on freshly isolated, single myofibers rarely expressed Mustn1(PRO) -GFP, but within 24 h of isolation, all activated satellite cells expressed Mustn1(PRO) -GFP. Expression of Mustn1(PRO) -GFP in primary myoblasts diminished with prolonged time in proliferation media. However, in response to serum withdrawal, the expression of Mustn1(PRO) -GFP increased during myofusion (day 2) followed by declining expression thereafter. CONCLUSION: Mustn1(PRO) -GFP is expressed in activated satellite cells and myoblasts but continued time in proliferation media diminished Mustn1(PRO) -GFP expression. However, myoblasts exposed to serum withdrawal increased Mustn1(PRO) -GFP expression consistent with its demonstrated role in myofusion. The in vivo expression pattern of Mustn1 observed in regenerating and developing skeletal muscle is consistent with its presence in satellite cells and its critical role in myofusion.


Assuntos
Proteínas de Fluorescência Verde/metabolismo , Músculo Esquelético/metabolismo , Mioblastos/citologia , Proteínas Nucleares/metabolismo , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Animais , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genes Reporter , Proteínas de Fluorescência Verde/genética , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Transgênicos , Músculo Esquelético/fisiologia , Mioblastos/fisiologia , Proteínas Nucleares/genética , Regiões Promotoras Genéticas
6.
Genesis ; 43(2): 87-98, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16149065

RESUMO

A 3.9 kb DNA fragment of human osteocalcin promoter and 3.6 kb DNA fragment of the rat collagen type1a1 promoter linked with visually distinguishable GFP isomers, topaz and cyan, were used for multiplex analysis of osteoblast lineage progression. Three patterns of dual transgene expression can be appreciated in primary bone cell cultures derived from the transgenic mice and by histology of their corresponding bones. Our data support the interpretation that strong pOBCol3.6GFPcyan alone is found in newly formed osteoblasts, while strong pOBCol3.6GFPcyan and hOC-GFPtpz are present in osteoblasts actively making a new matrix. Osteoblasts expressing strong hOC-GFPtpz and weak pOBCol3.6GFPcyan are also present and may or may not be producing mineralized matrix. This multiplex approach reveals the heterogeneity within the mature osteoblast population that cannot be appreciated by current histological methods. It should be useful to identify and isolate populations of cells within an osteoblast lineage as they progress through stages of differentiation.


Assuntos
Colágeno Tipo I/genética , Proteínas de Fluorescência Verde/análise , Substâncias Luminescentes/análise , Osteoblastos/química , Osteoblastos/citologia , Osteocalcina/genética , Animais , Diferenciação Celular , Células Cultivadas , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Ratos , Transgenes
7.
Bone ; 37(5): 678-87, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16112632

RESUMO

The interdependent relationship between vascular endothelial cells and osteoblasts during bone formation and fracture healing has been long appreciated. This paper reports a heterotopic implant model using FGF-2-expanded bone marrow stromal cells (BMSC) derived from Tie2eGFP (endothelial marker) and pOBCol3.6GFPcyan or topaz (early osteoblast marker) transgenic mice to appreciate the host/donor relationships of cells participating in the process of heterotopic bone formation. The study included various combinations of Tie2eGFP and pOBCol3.6GFPcyan and topaz transgenics as BMSC or whole bone marrow (WBM) donors and also as recipients. Rat tail collagen was used as a carrier of donor cells and implantation was done in lethally irradiated mice rescued with WBM injection. Development of ossicles in the implants was followed weekly during the 4- to 5-week long post-implantation period. By 4-5 weeks after total body irradiation (TBI) and implantation, a well-formed bone spicule had developed that was invested with bone marrow. Experiments showed absolute dominance of donor-derived cells in the formation of endothelial-lined vessels inside the implants as well as the marrow stromal-derived osteogenic cells. Host-derived fibroblasts and osteogenic cells were confined to the fibrous capsule surrounding the implant. In addition, cells lining the endosteal surface of newly formed marrow space carrying a pOBCol3.6GFP marker were observed that were contributed by WBM donor cells and the host. Thus, FGF-2-expanded BMSC appear to be a source of endothelial and osteogenic progenitor cells capable of eliciting heterotopic bone formation independent of cells from the host. This model should be useful for understanding the interactions between these two cell types that control osteogenic differentiation in vivo.


Assuntos
Transplante de Medula Óssea , Osso e Ossos/citologia , Colágeno , Endotélio Vascular/citologia , Osteoblastos/citologia , Osteogênese , Células Estromais/citologia , Animais , Biomarcadores , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Osso e Ossos/irrigação sanguínea , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/fisiologia , Diferenciação Celular , Endotélio Vascular/fisiologia , Géis , Implantes Experimentais , Camundongos , Camundongos Transgênicos , Osteoblastos/fisiologia , Radiografia , Células-Tronco/fisiologia , Células Estromais/fisiologia , Transplante Heterotópico , Irradiação Corporal Total
8.
Bone ; 35(1): 74-82, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15207743

RESUMO

Our previous studies have demonstrated that promoter-green fluorescent protein (GFP) transgenes can be used to identify and isolate populations of cells at the preosteoblastic stage (pOBCol3.6GFP) and at the mature osteoblastic stage (pOBCol2.3GFP) in living primary bone cell cultures. This strategy forms the basis for appreciating the cellular heterogeneity of lineage and relating gene function to cell differentiation. A weakness of this approach was the lack of a selective marker for late osteoblasts and mature osteocytes in the mineralized matrix. In this study, we have examined the expression of DMP-1 mRNA in murine marrow stromal and calvarial osteoblast cultures, and in bone, and calvaria in vivo. Furthermore, we have generated transgenic mice utilizing a mouse DMP1 cis-regulatory system to drive GFP as a marker for living osteocytes. Transgene expression was directed to mineralized tissues and showed a high correlation with the expression of the endogenous gene. Osteocyte-restricted expression of GFP was observed in histological sections of femur and calvaria and in primary cell cultures. Generation of this transgenic model will facilitate studies of gene expression and biological functions in these terminally differentiated bone cells.


Assuntos
Diferenciação Celular , Proteínas da Matriz Extracelular/biossíntese , Osteoblastos/metabolismo , Animais , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Linhagem da Célula , Células Cultivadas , Proteínas da Matriz Extracelular/genética , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Transgênicos , Osteoblastos/citologia , Osteócitos/citologia , Osteócitos/metabolismo , RNA Mensageiro/biossíntese , Células Estromais/citologia , Células Estromais/metabolismo
10.
Connect Tissue Res ; 43(2-3): 216-9, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12489162

RESUMO

Previous studies have shown that terminal differentiation of odontoblasts is accompanied by dramatic increases in type I collagen synthesis. Recently transgenic mice in which green fluorescent protein (GFP) expression is under the control of the rat 3.6 (pOBCol3.6GFPtpz) and 2.3 (pOBCol2.3GFPemd) Col1a1 promoter fragments were generated. Our analysis of these GFP-expressing transgenic mice shows that the 2.3-kb promoter fragment directs strong expression of GFP only to bones and teeth, whereas the 3.6-kb fragment of promoter directs strong expression of GFP in bone and tooth, as well as in other type I collagen producing tissues. Our observations of incisors in these transgenic mice show high levels of GFP expression in functional odontoblasts and in differentiated osteoblasts. These observations show that expression of GFP reporter genes closely follow the patterns of expression of alpha 1(I) collagen in various tissues including odontoblasts.


Assuntos
Colágeno Tipo I , Colágeno/genética , Expressão Gênica , Incisivo/fisiologia , Proteínas Luminescentes/genética , Transgenes , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Animais Recém-Nascidos/fisiologia , Diferenciação Celular , Colágeno/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Proteínas de Fluorescência Verde , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos/genética , Odontoblastos/metabolismo , Osteoblastos/metabolismo , Distribuição Tecidual
11.
Endocrinology ; 143(5): 1594-601, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11956140

RESUMO

This work examines the cellular pathophysiology associated with the weakened bone matrix found in a murine model of osteogenesis imperfecta murine (oim). Histomorphometric analysis of oim/oim bone showed significantly diminished bone mass, and the osteoblast and osteoclast histomorphometric parameters were increased in the oim/oim mice, compared with wild-type (+/+) mice. To assess osteoblast activity, a rat Col1a1 promoter linked to the chloramphenicol acetyltransferase reporter transgene was bred into the oim model. At 8 d and 1 month of age, no difference in transgene activity between oim and control mice was observed. However, at 3 months of age, chloramphenicol acetyl transferase activity was elevated in oim/oim;Tg/Tg, compared with +/+;Tg/Tg and oim/+;Tg/Tg. High levels of urinary pyridinoline crosslinks in the oim/oim;Tg/Tg mice were present at all ages, reflecting continuing high bone resorption. Our data portray a state of ineffective osteogenesis in which the mutant mouse never accumulates a normal quantity of bone matrix. However, it is only after the completion of the rapid growth phase that the high activity of the oim/oim osteoblast can compensate for the high rate of bone resorption. This relationship between bone formation and resorption may explain why the severity of osteogenesis imperfecta decreases after puberty is completed. The ability to quantify high bone turnover and advantages of using a transgene that reflects osteoblast lineage activity make this a useful model for studying interventions designed to improve the bone strength in osteogenesis imperfecta.


Assuntos
Matriz Óssea/fisiologia , Osteoblastos/fisiologia , Osteogênese Imperfeita/genética , Aminoácidos/urina , Animais , Biomarcadores/urina , Desenvolvimento Ósseo/fisiologia , Osso e Ossos/citologia , Cloranfenicol O-Acetiltransferase/biossíntese , Cloranfenicol O-Acetiltransferase/genética , Colágeno Tipo I/genética , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Tíbia/citologia
12.
Bone ; 31(6): 654-60, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12531558

RESUMO

The osteocalcin (OC) and a 2.3 kb fragment of the collagen promoter (Col2.3) have been used to restrict transgenic expression of a variety of proteins to bone. Transgenic mice carrying a green fluorescent protein (GFP) gene driven by each promoter were generated. Strong GFP expression was detected in OC-GFP mice in a few osteoblastic cells lining the endosteal bone surface and in scattered osteocytes within the bone matrix in long bones from 1-day-old to 6-month-old transgenic animals. Similar findings were noted in the forming tooth in which only individual odontoblasts expressed GFP without detectable expression from the dental pulp. This limited pattern of OC-GFP-positive cells contrasts with the uniform expression in the Col2.3GFP mice in which large proportion of osteoblasts, odontoblasts, and osteocytes strongly expressed the transgene. To assess transgene expression during in vitro differentiation, marrow stromal cell and neonatal calvarial osteoblast cultures were analyzed. The activity of both transgenes was restricted to mineralized nodules but the number of positive cells was lower in the OC-GFP-derived cultures. The different temporal and spatial pattern of each transgene in vivo and in vitro reveals potential advantages and disadvantages of these two transgene models.


Assuntos
Colágeno Tipo I/biossíntese , Proteínas Luminescentes/biossíntese , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteocalcina/biossíntese , Animais , Bovinos , Diferenciação Celular/fisiologia , Colágeno Tipo I/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Camundongos , Camundongos Transgênicos , Osteocalcina/genética , Regiões Promotoras Genéticas/fisiologia , Coelhos , Ratos , Transgenes/fisiologia , Células Tumorais Cultivadas
13.
Bone ; 29(4): 331-5, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11595615

RESUMO

This study tested the transduction efficiency of human bone marrow stromal cells (hBMSCs) with vesicular stomatitis virus (VSV)-pseudotyped retrovectors and their subsequent osteogenic differentiation in vitro. Two different retrovectors encoding beta-galactosidase (beta-gal) or enhanced green fluorescent protein (eGFP) as marker genes were examined for transduction of hBMSCs. hBMSCs were obtained from bone marrow filtrates of normal donors (aged 5-35 years), cultured in alpha-minimal essential medium (alpha-MEM) containing 10% fetal calf serum and infected with retrovectors soon after the adherent cells started to form individual colonies. Transduced hBMSCs were observed to express eGFP protein 4-7 days after infection in primary cultures, and the majority of hBMSCs were eGFP-positive. hBMSCs were also stained for beta-gal in the secondary cultures and virtually all hBMSCs expressed beta-gal activity. Transduced hBMSCs were examined for their osteogenic potential. These cells were found to express markers of osteogenic differentiation, including alkaline phosphatase, type I collagen, bone sialoprotein, decorin, and osteocalcin, as strongly as uninfected control cells. Mineralization was also induced by dexamethasone in transduced cells as well as control cells. These results demonstrate that hBMSCs are highly susceptible to infection with VSV-pseudotyped retrovectors with the majority of cultured cells expressing the viral transgenes without antibiotic selection. Transduced cells retain their osteogenic potential in vitro. hBMSCs are a promising cellular vehicle for systemic human gene therapy and VSV-pseudotyped retrovectors should be effective for their in vitro transduction prior to cellular engraftment.


Assuntos
Células da Medula Óssea/fisiologia , Vetores Genéticos , Osteoblastos/fisiologia , Células Estromais/fisiologia , Vírus da Estomatite Vesicular Indiana , Adolescente , Adulto , Células da Medula Óssea/citologia , Diferenciação Celular/genética , Criança , Pré-Escolar , Proteínas de Fluorescência Verde , Humanos , Indicadores e Reagentes/metabolismo , Proteínas Luminescentes/genética , Osteoblastos/citologia , Células Estromais/citologia , Transdução Genética/métodos , beta-Galactosidase/genética
14.
J Bone Miner Res ; 16(7): 1228-36, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11450698

RESUMO

The modular organization of the type I collagen promoter allows creation of promoter-reporter constructs with preferential activity in different type I collagen-producing tissues that might be useful to mark cells at different stages of osteoblastic differentiation. Primary marrow stromal cell (MSC) and mouse calvarial osteoblast (mCOB) cultures were established from transgenic mice harboring different Col1a1 promoter fragments driving chloramphenicol acetyltransferase (CAT). In these models, Col1a1 messenger RNA (mRNA) and alkaline phosphatase (ALP) are the first markers of differentiation appearing soon after the colonies develop. Bone sialoprotein (BSP) is detected 2-3 days later, followed by osteocalcin (OC) expression and nodule mineralization. A 3.6 Col1a1 fragment (ColCAT3.6) initiated activity concomitant with ALP staining and type I collagen mRNA expression. In contrast, a 2.3 Col1a1 fragment (ColCAT2.3) became active coincident with BSP expression. The pattern of transgene expression assessed by immunostaining was distinctly different. ColCAT3.6 was expressed within and at the periphery of developing nodules whereas the ColCAT2.3 expression was restricted to the differentiated nodules. The feasibility of using green fluorescent protein (GFP) as a marker of osteoblast differentiation was evaluated in ROS17/2.8 cells. A 2.3-kilobase (kb) Col1a1 promoter driving GFP (pOB4Col2.3GLP) was stably transfected into the cell line and positive clones were selected. Subcultures lost and then regained GFP expression that was localized in small clusters of cells throughout the culture. This suggests that expression from the 2.3-kb Col1A1 fragment is determined by the state of differentiation of the ROS17/2.8 cells. Col1a1 transgenes should be useful in appreciating the heterogeneity of a primary or immortalized culture undergoing osteoblastic differentiation.


Assuntos
Linhagem da Célula/genética , Colágeno Tipo I , Colágeno/genética , Osteoblastos/citologia , Regiões Promotoras Genéticas/genética , Transgenes/genética , Animais , Diferenciação Celular/genética , Células Cultivadas , Cloranfenicol O-Acetiltransferase/genética , Cloranfenicol O-Acetiltransferase/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Genes Reporter/genética , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Osteoblastos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Células Estromais/citologia , Células Estromais/metabolismo , Transfecção , Células Tumorais Cultivadas
15.
Croat Med J ; 42(4): 393-415, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11471191

RESUMO

Osteogenesis imperfecta (OI), or brittle bone disease, is a heritable disorder characterized by increased bone fragility. Four different types of the disease are commonly distinguished, ranging from a mild condition (type I) to a lethal one (type II). Types III and IV are the severe forms surviving the neonatal period. In most cases, there is a reduction in the production of normal type I collagen or the synthesis of abnormal collagen as a result of mutations in the type I collagen genes. These classic forms of OI are described in this review. There are instances, however, where alterations in bone matrix components, other than type I collagen, are the basic abnormalities of the OI. Recently, three such discrete types have been identified by histomorphometric evaluation (types V and VI) and linkage analysis (Rhizomelic OI). They provide evidence for the as yet poorly understood complexity of the phenotype-genotype correlation in OI. We also discuss bisphosphonates treatment as well as fracture management and surgical correction of deformities observed in the patients with OI. However, ultimately, strengthening bone in OI will involve steps to correct the underlying genetic mutations that are responsible for this disorder. Thus, we also describe different genetic therapeutic approaches that have been tested either on OI cells or on available OI murine models.


Assuntos
Osteogênese Imperfeita/genética , Animais , Criança , Difosfonatos/uso terapêutico , Modelos Animais de Doenças , Ligação Genética , Terapia Genética , Humanos , Camundongos , Mutação , Osteogênese Imperfeita/classificação , Osteogênese Imperfeita/complicações , Osteogênese Imperfeita/fisiopatologia , Osteogênese Imperfeita/terapia , Platibasia/etiologia , Platibasia/fisiopatologia , Polimorfismo Conformacional de Fita Simples , Escoliose/etiologia
16.
Croat Med J ; 42(4): 436-9, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11471195

RESUMO

AIM: To determine whether retrovirally expressed Dlx5, a homeobox-containing transcription factor, can induce a 2.3 kb rat COL1A1 promoter-reporter construct, which is transduced into osteoblastic cells by the use of a retrovirus vector. METHODS: A self-inactivating retrovirus vector containing the rat COL1A1 driving green fluorescent protein (GFP) was transduced into chick calvarial periosteal cells. These cells were then infected with a replication-competent retroviral vector expressing Dlx5, or a control vector. The cells were cultured in the presence of ascorbic acid and beta-glycerol-phosphate, which promotes osteoblastic differentiation. Expression of the COL1A1 promoter was assessed by detecting GFP with fluorescence microscopy. RESULTS: GFP was detected only in cells infected with the Dlx5 expressing retrovirus. The GFP positive cells were observed in regions of the culture that had undergone osteoblastic differentiation, as detected by cell morphology and the presence of a mineralized matrix. CONCLUSION: The 2.3 kb rat COL1A1 promoter fragment contains elements responsive to Dlx5, and the self-inactivating retroviral vector allows these elements to be used appropriately.


Assuntos
Colágeno Tipo I , Colágeno/metabolismo , Proteínas de Homeodomínio/fisiologia , Retroviridae/genética , Animais , Diferenciação Celular , Células Cultivadas , Embrião de Galinha , Cadeia alfa 1 do Colágeno Tipo I , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Ratos , Transfecção
17.
Virology ; 284(1): 37-45, 2001 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-11352666

RESUMO

Marrow stromal cells (MSC) and neonatal calvarial cells have the potential to differentiate and express markers of mature osteoblasts. Furthermore, MSCs can generate multiple differentiated connective tissue phenotypes. These properties and their ability to be expanded ex vivo make them good models for ex vivo gene therapy. In this study we examined the ability of vesicular stomatitis virus (VSV-G) pseudotyped retroviral vectors to transduce osteoprogenitor cells derived from bone marrow and from neonatal calvaria. Retrovectors encoding either beta-galactosidase or green fluorescent protein (eGFP) were used for transduction of primary murine marrow stromal and primary neonatal calvarial cell cultures. High infection efficiency was demonstrated by fluorescence-activated cell analysis when GFP was used as a marker or by estimating the number of beta-galactosidase-positive cells. Expression of markers of differentiated bone cells, including Col1a1, bone sialoprotein, and osteocalcin mRNA and alkaline phosphatase activity was not impaired by retroviral transduction. Our data suggest that VSV-G pseudotypes retroviral vectors are suitable for introducing genes into osteoprogenitor cells without affecting osteoprogenitor lineage progression.


Assuntos
Glicoproteínas de Membrana , Osteoblastos/virologia , Retroviridae , Transformação Genética , Proteínas do Envelope Viral/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Vetores Genéticos , Proteínas de Fluorescência Verde , Proteínas Luminescentes/metabolismo , Camundongos
18.
Mol Cell Biol ; 21(8): 2815-25, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11283260

RESUMO

Although the primary function of U1 snRNA is to define the 5' donor site of an intron, it can also block the accumulation of a specific RNA transcript when it binds to a donor sequence within its terminal exon. This work was initiated to investigate if this property of U1 snRNA could be exploited as an effective method for inactivating any target gene. The initial 10-bp segment of U1 snRNA, which is complementary to the 5' donor sequence, was modified to recognize various target mRNAs (chloramphenicol acetyltransferase [CAT], beta-galactosidase, or green fluorescent protein [GFP]). Transient cotransfection of reporter genes and appropriate U1 antitarget vectors resulted in >90% reduction of transgene expression. Numerous sites within the CAT transcript were suitable for targeting. The inhibitory effect of the U1 antitarget vector is directly related to the hybrid formed between the U1 vector and target transcripts and is dependent on an intact 70,000-molecular-weight binding domain within the U1 gene. The effect is long lasting when the target (CAT or GFP) and U1 antitarget construct are inserted into fibroblasts by stable transfection. Clonal cell lines derived from stable transfection with a pOB4GFP target construct and subsequently stably transfected with the U1 anti-GFP construct were selected. The degree to which GFP fluorescence was inhibited by U1 anti-GFP in the various clonal cell lines was assessed by fluorescence-activated cell sorter analysis. RNA analysis demonstrated reduction of the GFP mRNA in the nuclear and cytoplasmic compartment and proper 3' cleavage of the GFP residual transcript. An RNase protection strategy demonstrated that the transfected U1 antitarget RNA level varied between 1 to 8% of the endogenous U1 snRNA level. U1 antitarget vectors were demonstrated to have potential as effective inhibitors of gene expression in intact cells.


Assuntos
Expressão Gênica , Marcação de Genes/métodos , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/metabolismo , Células 3T3 , Animais , Sequência de Bases , Cloranfenicol O-Acetiltransferase/genética , DNA/genética , Genes Reporter , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Camundongos , Transfecção , beta-Galactosidase/genética
19.
Mol Ther ; 3(4): 543-50, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11319916

RESUMO

Gene therapy of bone would benefit from the availability of vectors that provide stable, osteoblast-specific expression. This would allow bone-specific expression of Col1a1 cDNAs for treatment of osteogenesis imperfecta. In addition, such a vector would restrict expression of secreted therapeutic proteins to the bone-synthesizing regions of the bone marrow after ex vivo transduction of marrow stromal cells and reintroduction of the cells into patients. Retrovirus vectors stably integrate into target cell genomes; however, long-term regulated expression from internal cellular promoters has not been consistently achieved. In some cases this is due to a stem cell-specific mechanism for transcriptional repression of retroviruses. We evaluated the ability of self-inactivating ROSA-derived vectors containing a bone-directed 2.3-kb rat Col1a1 promoter to display osteoblast-specific expression. In vitro expression was examined in bone marrow stromal cell cultures induced to undergo osteoblastic differentiation. In vivo expression was evaluated in chimeric mice derived from transduced embryonic stem cells. The results indicate that self-inactivating retrovirus vectors containing the Col1a1 promoter are not permanently inactivated in embryonic stem cells and are specifically expressed in osteoblasts in vivo and in vitro. Thus these vectors should be useful for bone-directed gene therapy.


Assuntos
Células da Medula Óssea/citologia , Osso e Ossos/metabolismo , Colágeno Tipo I , Colágeno/genética , Camundongos Transgênicos , Regiões Promotoras Genéticas , Retroviridae/genética , Animais , Linhagem Celular , Células Cultivadas , Cadeia alfa 1 do Colágeno Tipo I , DNA Complementar/metabolismo , Embrião de Mamíferos/citologia , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/biossíntese , Camundongos , Microscopia de Contraste de Fase , Modelos Genéticos , Osteoblastos/metabolismo , Ratos , Células-Tronco/metabolismo , Fatores de Tempo , Transdução Genética , Transfecção , beta-Galactosidase/metabolismo
20.
J Bone Miner Res ; 16(12): 2222-31, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11760835

RESUMO

Two transgenic mouse lines were generated with a DNA construct bearing a 2.3-kilobase (kb) fragment of the rat alpha1 type I collagen promoter driving a truncated form of the herpes thymidine kinase gene (Col2.3Atk). Expression of the transgene was found in osteoblasts coincident with other genetic markers of early osteoblast differentiation. Mice treated with ganciclovir (GCV) for 16 days displayed extensive destruction of the bone lining cells and decreased osteoclast number. In addition, a dramatic decrease in bone marrow elements was observed, which was more severe in the primary spongiosum and marrow adjacent to the diaphyseal endosteal bone. Immunostaining for transgene expression within the bone marrow was negative and marrow stromal cell cultures developed normally in the presence of GCV until the point of early osteoblast differentiation. Our findings suggest that the early differentiating osteoblasts are necessary for the maintenance of osteoclasts and hematopoiesis. Termination of GCV treatment produced an exaggerated response of new bone formation in cortical and trabecular bone. The Col2.3deltatk mouse should be a useful model to define the interrelation between bone and marrow elements as well as a model to analyze the molecular and cellular events associated with a defined wave of osteogenesis on termination of GCV treatment.


Assuntos
Osteoblastos/citologia , Fosfatase Alcalina/metabolismo , Animais , Biomarcadores , Diferenciação Celular , Linhagem da Célula , Chlorocebus aethiops , Colágeno Tipo I/genética , Ganciclovir/farmacologia , Expressão Gênica , Camundongos , Camundongos Transgênicos , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Ratos , Sialoglicoproteínas/metabolismo , Simplexvirus/enzimologia , Simplexvirus/genética , Timidina Quinase/genética , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...