Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Regen Ther ; 14: 19-31, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31988991

RESUMO

Clinically relevant human induced pluripotent stem cell (hiPSC) derivatives require efficient protocols to differentiate hiPSCs into specific lineages. Here we developed a fully defined xeno-free strategy to direct hiPSCs toward osteoblasts within 21 days. The strategy successfully achieved the osteogenic induction of four independently derived hiPSC lines by a sequential use of combinations of small-molecule inducers. The induction first generated mesodermal cells, which subsequently recapitulated the developmental expression pattern of major osteoblast genes and proteins. Importantly, Col2.3-Cherry hiPSCs subjected to this strategy strongly expressed the cherry fluorescence that has been observed in bone-forming osteoblasts in vivo. Moreover, the protocol combined with a three-dimensional (3D) scaffold was suitable for the generation of a xeno-free 3D osteogenic system. Thus, our strategy offers a platform with significant advantages for bone biology studies and it will also contribute to clinical applications of hiPSCs to skeletal regenerative medicine.

2.
J Histochem Cytochem ; 67(6): 401-417, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30848692

RESUMO

The effectiveness of autologous cell-based skeletal repair continues to be controversial in part because in vitro predictors of in vivo human bone formation by cultured human progenitor cells are not reliable. To assist in the development of in vivo assays of human osteoprogenitor potential, a fluorescence-based histology of nondecalcified mineralized tissue is presented that provides multiple criteria to distinguish human and host osteoblasts, osteocytes, and accumulated bone matrix in a mouse calvarial defect model. These include detection of an ubiquitously expressed red fluorescent protein reporter by the implanted human cells, antibodies specific to human bone sialoprotein and a human nuclear antigen, and expression of a bone/fibroblast restricted green fluorescent protein reporter in the host tissue. Using low passage bone marrow-derived stromal cells, robust human bone matrix formation was obtained. However, a striking feature is the lack of mouse bone marrow investment and osteoclasts within the human bone matrix. This deficiency may account for the accumulation of a disorganized human bone matrix that has not undergone extensive remodeling. These features, which would not be appreciated by traditional decalcified paraffin histology, indicate the human bone matrix is not undergoing active remodeling and thus the full differentiation potential of the implanted human cells within currently used mouse models is not being realized.


Assuntos
Osteogênese , Animais , Humanos , Camundongos , Imagem Óptica , Osteoblastos/citologia , Crânio/citologia , Crânio/diagnóstico por imagem , Crânio/fisiologia
3.
Int J Nanomedicine ; 13: 3937-3948, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30013346

RESUMO

BACKGROUND: Intravenously (IV)-injected gold nanoparticles (AuNPs) powerfully enhance the efficacy of X-ray therapy of tumors including advanced gliomas. However, pharmacokinetic issues, such as slow tissue clearance and skin discoloration, may impede clinical translation. The direct infusion of AuNPs into the tumor might be an alternative mode of delivery. MATERIALS AND METHODS: Using the advanced, invasive, and difficult-to-treat F98 rat glioma model, we have studied the biodistribution of the AuNPs in the tumor and surrounding brain after either IV injection or direct intratumoral infusion by convection-enhanced delivery using light microscopy immunofluorescence and direct gold visualization. RESULTS: IV-injected AuNPs localize more specifically to intracerebral tumor cells, both in the main tumor mass and in the migrated tumor cells as well as the tumor edema, than do the directly infused AuNPs. Although some of the directly infused AuNPs do access the main tumor region, such access is largely restricted. CONCLUSION: These data suggest that IV-injected AuNPs are likely to have a greater therapeutic benefit when combined with radiation therapy than after the direct infusion of AuNPs.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Convecção , Sistemas de Liberação de Medicamentos , Glioma/tratamento farmacológico , Ouro/química , Nanopartículas Metálicas/química , Animais , Astrócitos/patologia , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Edema/patologia , Glioma/irrigação sanguínea , Glioma/patologia , Injeções Intravenosas , Microglia/patologia , Invasividade Neoplásica , Ratos , Ratos Endogâmicos F344 , Técnicas Estereotáxicas , Distribuição Tecidual
4.
Nat Commun ; 9(1): 300, 2018 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-29352112

RESUMO

Bone homeostasis is regulated by communication between bone-forming mature osteoblasts (mOBs) and bone-resorptive mature osteoclasts (mOCs). However, the spatial-temporal relationship and mode of interaction in vivo remain elusive. Here we show, by using an intravital imaging technique, that mOB and mOC functions are regulated via direct cell-cell contact between these cell types. The mOBs and mOCs mainly occupy discrete territories in the steady state, although direct cell-cell contact is detected in spatiotemporally limited areas. In addition, a pH-sensing fluorescence probe reveals that mOCs secrete protons for bone resorption when they are not in contact with mOBs, whereas mOCs contacting mOBs are non-resorptive, suggesting that mOBs can inhibit bone resorption by direct contact. Intermittent administration of parathyroid hormone causes bone anabolic effects, which lead to a mixed distribution of mOBs and mOCs, and increase cell-cell contact. This study reveals spatiotemporal intercellular interactions between mOBs and mOCs affecting bone homeostasis in vivo.


Assuntos
Reabsorção Óssea/diagnóstico por imagem , Comunicação Celular/fisiologia , Osteoblastos/citologia , Osteoclastos/citologia , Osteogênese/fisiologia , Animais , Diferenciação Celular , Feminino , Corantes Fluorescentes/química , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Homeostase/fisiologia , Concentração de Íons de Hidrogênio , Microscopia Intravital/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoblastos/efeitos dos fármacos , Osteoblastos/fisiologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/fisiologia , Hormônio Paratireóideo/farmacologia , Cultura Primária de Células , Crânio/citologia , Crânio/diagnóstico por imagem , Crânio/efeitos dos fármacos , Crânio/fisiologia
5.
Methods Mol Biol ; 1723: 385-396, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29344873

RESUMO

Laser-capture microdissection (LCM) coupled to downstream RNA analysis poses unique difficulties for the evaluation of mineralized tissues. A rapid protocol was thus developed to enable sufficient integrity of bone and cartilage tissue for reliable sectioning, while minimizing RNA loss associated with prolonged decalcification and purification steps. Specifically, the protocol involves pump-assisted, cardiac perfusion-fixation with paraformaldehyde, and moderate digestion of LCM-acquired tissue with proteinase K followed by DNase treatment and separation of RNA using magnetic beads. Reverse transcription and cDNA synthesis are performed immediately after RNA purification, without need for further protein removal.


Assuntos
Osso e Ossos/metabolismo , Cartilagem/metabolismo , Modelos Animais de Doenças , Células-Tronco Pluripotentes Induzidas/metabolismo , Microdissecção e Captura a Laser/métodos , Células-Tronco Mesenquimais/metabolismo , RNA/análise , Crânio/metabolismo , Animais , Osso e Ossos/patologia , Cartilagem/patologia , Perfilação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Mesenquimais/patologia , Camundongos , Perfusão , RNA/genética , RNA/isolamento & purificação , Crânio/patologia
6.
Sci Adv ; 3(5): e1602875, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28508073

RESUMO

The development of in vitro models for the maintenance and differentiation of pluripotent stem cells (PSCs) is an active area of stem cell research. The strategies used so far are based mainly on two-dimensional (2D) cultures, in which cellular phenotypes are regulated by soluble factors. We show that a 3D culture system with atelocollagen porous scaffolds can significantly improve the outcome of the current platforms intended for the maintenance and lineage specification of mouse PSCs (mPSCs). Unlike 2D conditions, the 3D conditions maintained the undifferentiated state of mouse embryonic stem cells (mESCs) without exogenous stimulation and also supported endoderm, mesoderm, and ectoderm differentiation of mESCs under serum-free conditions. Moreover, 3D mPSC-derived mesodermal cells showed accelerated osteogenic differentiation, giving rise to functional osteoblast-osteocyte populations within calcified structures. The present strategy offers a 3D platform suitable for the formation of organoids that mimic in vivo organs containing various cell types, and it may be adaptable to the generation of ectoderm-, mesoderm-, and endoderm-derived tissues when combined with appropriate differentiation treatments.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Osteócitos/metabolismo , Osteogênese , Células-Tronco Pluripotentes/metabolismo , Alicerces Teciduais/química , Animais , Camundongos , Osteócitos/citologia , Células-Tronco Pluripotentes/citologia
7.
Stem Cells Transl Med ; 3(10): 1125-37, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25122686

RESUMO

The use of human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) for study and treatment of bone diseases or traumatic bone injuries requires efficient protocols to differentiate hESCs/iPSCs into cells with osteogenic potential and the ability to isolate differentiated osteoblasts for analysis. We have used zinc finger nuclease technology to deliver a construct containing the Col2.3 promoter driving GFPemerald to the AAVS1 site (referred to as a "safe harbor" site), in human embryonic stem cells (H9Zn2.3GFP), with the goal of marking the cells that have become differentiated osteoblasts. In teratomas formed using these cells, we identified green fluorescent protein (GFP)-positive cells specifically associated with in vivo bone formation. We also differentiated the cells into a mesenchymal stem cell population with osteogenic potential and implanted them into a mouse calvarial defect model. We observed GFP-positive cells associated with alizarin complexone-labeled newly formed bone surfaces. The cells were alkaline phosphatase-positive, and immunohistochemistry with human specific bone sialoprotein (BSP) antibody indicates that the GFP-positive cells are also associated with the human BSP-containing matrix, demonstrating that the Col2.3GFP construct marks cells in the osteoblast lineage. Single-cell cloning generated a 100% Col2.3GFP-positive cell population, as demonstrated by fluorescence in situ hybridization using a GFP probe. The karyotype was normal, and pluripotency was demonstrated by Tra1-60 immunostaining, pluripotent low density reverse transcription-polymerase chain reaction array and embryoid body formation. These cells will be useful to develop optimal osteogenic differentiation protocols and to isolate osteoblasts from normal and diseased iPSCs for analysis.


Assuntos
Células-Tronco Embrionárias/citologia , Proteínas de Fluorescência Verde , Osteoblastos/citologia , Animais , Matriz Óssea/citologia , Diferenciação Celular/fisiologia , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Genes Reporter , Xenoenxertos , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Camundongos , Células-Tronco Pluripotentes/citologia , Ratos
8.
Stem Cell Reports ; 2(6): 751-60, 2014 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-24936463

RESUMO

Pluripotent stem cells are a promising tool for mechanistic studies of tissue development, drug screening, and cell-based therapies. Here, we report an effective and mass-producing strategy for the stepwise differentiation of mouse embryonic stem cells (mESCs) and mouse and human induced pluripotent stem cells (miPSCs and hiPSCs, respectively) into osteoblasts using four small molecules (CHIR99021 [CHIR], cyclopamine [Cyc], smoothened agonist [SAG], and a helioxanthin-derivative 4-(4-methoxyphenyl)pyrido[4',3':4,5]thieno[2,3-b]pyridine-2-carboxamide [TH]) under serum-free and feeder-free conditions. The strategy, which consists of mesoderm induction, osteoblast induction, and osteoblast maturation phases, significantly induced expressions of osteoblast-related genes and proteins in mESCs, miPSCs, and hiPSCs. In addition, when mESCs defective in runt-related transcription factor 2 (Runx2), a master regulator of osteogenesis, were cultured by the strategy, they molecularly recapitulated osteoblast phenotypes of Runx2 null mice. The present strategy will be a platform for biological and pathological studies of osteoblast development, screening of bone-augmentation drugs, and skeletal regeneration.


Assuntos
Técnicas de Cultura de Células/métodos , Meios de Cultura Livres de Soro/farmacologia , Osteoblastos/citologia , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Cicloexilaminas/farmacologia , Humanos , Camundongos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Piridinas/farmacologia , Pirimidinas/farmacologia , Tiofenos/farmacologia , Alcaloides de Veratrum/farmacologia
9.
Artigo em Inglês | MEDLINE | ID: mdl-25570850

RESUMO

Bone defect repair is a significant clinical challenge in orthopedic surgery. Despite tremendous efforts, the majority of the current bone tissue engineering strategies depend on bone formation via intramembranous ossification (IO), which often results in poor vascularization and limited-area bone regeneration. Recently, there has been increasing interest in exploring bone regeneration through a cartilage-mediated process similar to endochondral ossification (EO). This method is advantageous because long bones are originally developed through EO and moreover, vascularization is an inherent step of this process. Therefore, it may be possible to effectively employ the EO method for the repair and regeneration of large and segmental bone defects. Although a number of studies have demonstrated engineered bone formation through EO, there are no approaches aiming for their clinical translation. In this study, we propose a strategy modeled after the U.S. Food and Drug Administration (FDA) approved autologus chondrocyte implantation (ACI) procedure. In its implementation, we concentrated human bone marrow aspirate via a minimally manipulated process and demonstrated the potential of human bone marrow derived cells for in vitro pre-cartilage template formation and bone regeneration in vivo.


Assuntos
Osso e Ossos/fisiologia , Engenharia Tecidual , Adolescente , Adulto , Animais , Células da Medula Óssea/citologia , Regeneração Óssea , Células Cultivadas , Condrócitos/citologia , Condrócitos/metabolismo , Feminino , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Transgênicos , Osteogênese , Radiografia , Crânio/diagnóstico por imagem , Crânio/patologia , Transplante Heterólogo , Cicatrização , Adulto Jovem
10.
Tissue Eng Part A ; 20(1-2): 365-77, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23952622

RESUMO

The in vivo osteogenesis potential of mesenchymal-like cells derived from human embryonic stem cells (hESC-MCs) was evaluated in vivo by implantation on collagen/hydroxyapatite scaffolds into calvarial defects in immunodeficient mice. This study is novel because no osteogenic or chondrogenic differentiation protocols were applied to the cells prior to implantation. After 6 weeks, X-ray, microCT, and histological analysis showed that the hESC-MCs had consistently formed a highly vascularized new bone that bridged the bone defect and seamlessly integrated with host bone. The implanted hESC-MCs differentiated in situ to functional hypertrophic chondrocytes, osteoblasts, and osteocytes forming new bone tissue via an endochondral ossification pathway. Evidence for the direct participation of the human cells in bone morphogenesis was verified by two separate assays: with Alu and by human mitochondrial antigen positive staining in conjunction with co-localized expression of human bone sialoprotein in histologically verified regions of new bone. The large volume of new bone in a calvarial defect and the direct participation of the hESC-MCs far exceeds that of previous studies and that of the control adult hMSCs. This study represents a key step forward for bone tissue engineering because of the large volume, vascularity, and reproducibility of new bone formation and the discovery that it is advantageous to not over-commit these progenitor cells to a particular lineage prior to implantation. The hESC-MCs were able to recapitulate the mesenchymal developmental pathway and were able to repair the bone defect semi-autonomously without preimplantation differentiation to osteo- or chondroprogenitors.


Assuntos
Regeneração Óssea/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Mesenquimais/citologia , Adulto , Animais , Regeneração Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Colágeno/farmacologia , Durapatita/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Morfogênese/efeitos dos fármacos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , Fenótipo , Implantação de Prótese , Regeneração/efeitos dos fármacos , Crânio/efeitos dos fármacos , Crânio/patologia , Transplante de Células-Tronco , Alicerces Teciduais/química
11.
Cell Reprogram ; 15(6): 503-13, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24219578

RESUMO

Studies of rare genetic bone disorders are often limited due to unavailability of tissue specimens and the lack of animal models fully replicating phenotypic features. Craniometaphyseal dysplasia (CMD) is a rare monogenic disorder characterized by hyperostosis of craniofacial bones concurrent with abnormal shape of long bones. Mutations for autosomal dominant CMD have been identified in the ANK gene (ANKH). Here we describe a simple and efficient method to reprogram adherent cells cultured from peripheral blood to human induced pluripotent stem cells (hiPSCs) from eight CMD patients and five healthy controls. Peripheral blood mononuclear cells (PBMCs) were separated from 5-7 mL of whole blood by Ficoll gradient, expanded in the presence of cytokines and transduced with Sendai virus (SeV) vectors encoding OCT3/4, SOX2, KLF4, and c-MYC. SeV vector, a cytoplasmic RNA vector, is lost from host cells after propagation for 10-13 passages. These hiPSCs express stem cell markers, have normal karyotypes, and are capable of forming embryoid bodies in vitro as well as teratomas in vivo. Further differentiation of these patient-specific iPSCs into osteoblasts and osteoclasts can provide a useful tool to study the effects CMD mutations on bone, and this approach can be applied for disease modeling of other rare genetic musculoskeletal disorders.


Assuntos
Doenças do Desenvolvimento Ósseo/sangue , Reprogramação Celular , Anormalidades Craniofaciais/sangue , Vetores Genéticos , Hiperostose/sangue , Hipertelorismo/sangue , Células-Tronco Pluripotentes Induzidas/citologia , Vírus Sendai/genética , Adulto , Sequência de Bases , Estudos de Casos e Controles , Criança , Primers do DNA , Feminino , Rearranjo Gênico do Linfócito T , Humanos , Fator 4 Semelhante a Kruppel , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase
12.
Connect Tissue Res ; 54(4-5): 296-304, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23782451

RESUMO

Previous studies reported that embryonic stem cells (ESCs) can be induced to differentiate into cells showing a mature osteoblastic phenotype by culturing them under osteo-inductive conditions. It is probable that osteogenic differentiation requires that ESCs undergo differentiation through an intermediary step involving a mesenchymal lineage precursor. Based on our previous studies indicating that adult mesenchymal progenitor cells express α-smooth muscle actin (αSMA), we have generated ESCs from transgenic mice in which an αSMA promoter directs the expression of red fluorescent protein (RFP) to mesenchymal progenitor cells. To track the transition of ESC-derived MSCs into mature osteoblasts, we have utilized a bone-specific fragment of rat type I collagen promoter driving green fluorescent protein (Col2.3GFP). Following osteogenic induction in ESCs, we have observed expression of alkaline phosphatase (ALP) and subsequent mineralization as detected by von Kossa staining. After 1 week of osteogenic induction, ESCs begin to express αSMARFP. This expression was localized to the peripheral area encircling a typical ESC colony. Nevertheless, these αSMARFP positive cells did not show activation of the Col2.3GFP promoter, even after 7 weeks of osteogenic differentiation in vitro. In contrast, Col2.3GFP expression was detected in vivo, in mineralized areas following teratoma formation. Our results indicate that detection of ALP activity and mineralization of ESCs cultured under osteogenic conditions is not sufficient to demonstrate osteogenic maturation. Our study indicates the utility of the promoter-visual transgene approach to assess the commitment and differentiation of ESCs into the osteoblast lineage.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Osteogênese/fisiologia , Fosfatase Alcalina/metabolismo , Animais , Camundongos , Camundongos Transgênicos
13.
PLoS One ; 5(7): e11853, 2010 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-20686615

RESUMO

BACKGROUND: Directed differentiation of human induced pluripotent stem cells (hiPSC) into functional, region-specific neural cells is a key step to realizing their therapeutic promise to treat various neural disorders, which awaits detailed elucidation. METHODOLOGY/PRINCIPAL FINDINGS: We analyzed neural differentiation from various hiPSC lines generated by others and ourselves. Although heterogeneity in efficiency of neuroepithelial (NE) cell differentiation was observed among different hiPSC lines, the NE differentiation process resembles that from human embryonic stem cells (hESC) in morphology, timing, transcriptional profile, and requirement for FGF signaling. NE cells differentiated from hiPSC, like those from hESC, can also form rostral phenotypes by default, and form the midbrain or spinal progenitors upon caudalization by morphogens. The rostrocaudal neural progenitors can further mature to develop forebrain glutamatergic projection neurons, midbrain dopaminergic neurons, and spinal motor neurons, respectively. Typical ion channels and action potentials were recorded in the hiPSC-derived neurons. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that hiPSC, regardless of how they were derived, can differentiate into a spectrum of rostrocaudal neurons with functionality, which supports the considerable value of hiPSC for study and treatment of patient-specific neural disorders.


Assuntos
Diferenciação Celular/fisiologia , Ácido Glutâmico/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Neurônios/citologia , Neurônios/metabolismo , Prosencéfalo/citologia , Diferenciação Celular/genética , Eletrofisiologia , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Coll Antropol ; 33(3): 919-24, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19860126

RESUMO

We analyzed proliferation and differentiation of calvarial osteoblasts derived from Msx2 deficient in comparison with wild type mice. Calvarial osteoblast cultures from five to eight days old Msx2 deficient, heterozygous and wild type mice were studied for difference in proliferation and differentiation. Proliferation rate was assessed by counting cell number, BrdU and Calcein AM labeling. Differentiation was assessed by Von Kossa and alkaline phosphatase staining, northern blot hybridization with bone differentiation markers, infection of cell cultures with retrovirus expressing GFP under the control of type I collagen promoter fragment. At day six, cell number in cell culture derived from Msx2 deficient mice was 20% lower then in culture from wild type mice. There were 16.8% BrdU labeled cells in cell culture from Msx2 deficient mice, 20.9% in culture from heterozygous mice and 21.6% in culture from wild type mice. Cell cultures from Msx2 deficient mice showed lower intensity of fluorescence when marked with Calcein AM then cultures from wild type mice. Von Kossa staining showed increased mineralization and northern blot analysis showed increased levels of bone differentiation markers in cell cultures derived from Msx2 deficient mice. GFP came on earlier in Msx2 deficient cultures after infection with Col2.3 GFP retrovirus. We conclude that calvarial osteoblasts derived from Msx2 deficient mice have a lower rate of proliferation and demonstrate increased osteoblastic differentiation when compared to osteoblasts derived from wild type mice.


Assuntos
Proteínas de Homeodomínio/fisiologia , Osteoblastos/citologia , Crânio/crescimento & desenvolvimento , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
15.
Mol Ther ; 17(11): 1967-78, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19603005

RESUMO

Bone marrow transplantation can provide an effective cell-based strategy to enhance bone repair. However, the fate of implanted cells and the extent of their contribution to bone osteoinduction remain uncertain. To define the fate of bone marrow-derived cells and their contribution in vivo, we used a bone-specific collagen I promoter (2.3Col) driving green fluorescent protein (GFP) (2.3ColGFP) within a lentiviral vector. Prior to in vivo cell fate determination, we verified a high efficiency of lentiviral transduction in human bone marrow stromal cells (hBMSCs), without altering the proliferation or differentiation potential of these cells. We showed that the 2.3ColGFP marker responded to endogenous transcriptional regulation signals. In a mouse ossicle model, we demonstrated that the 2.3ColGFP marker is able to specifically define human bone marrow-derived stem cells that enter the osteoblast lineage in vivo. In addition, cells labeled with 2.3ColGFP with the donor origin, directly make a major contribution to bone formation. Furthermore, we also demonstrated in a calvarial defect model that a mixture of human bone marrow-derived populations, have stronger bone regenerative potential than that of hBMSCs, and an optimal dose is required for bone regeneration by the mixed populations.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Osteogênese/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Colágeno Tipo I/genética , Citometria de Fluxo , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Humanos , Lentivirus/genética , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
BMC Biotechnol ; 9: 20, 2009 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-19284652

RESUMO

BACKGROUND: Reporter gene mice are valuable animal models for biological research providing a gene expression readout that can contribute to cellular characterization within the context of a developmental process. With the advancement of bacterial recombination techniques to engineer reporter gene constructs from BAC genomic clones and the generation of optically distinguishable fluorescent protein reporter genes, there is an unprecedented capability to engineer more informative transgenic reporter mouse models relative to what has been traditionally available. RESULTS: We demonstrate here our first effort on the development of a three stage bacterial recombination strategy to physically link multiple genes together with their respective fluorescent protein (FP) reporters in one DNA fragment. This strategy uses bacterial recombination techniques to: (1) subclone genes of interest into BAC linking vectors, (2) insert desired reporter genes into respective genes and (3) link different gene-reporters together. As proof of concept, we have generated a single DNA fragment containing the genes Trap, Dmp1, and Ibsp driving the expression of ECFP, mCherry, and Topaz FP reporter genes, respectively. Using this DNA construct, we have successfully generated transgenic reporter mice that retain two to three gene readouts. CONCLUSION: The three stage methodology to link multiple genes with their respective fluorescent protein reporter works with reasonable efficiency. Moreover, gene linkage allows for their common chromosomal integration into a single locus. However, the testing of this multi-reporter DNA construct by transgenesis does suggest that the linkage of two different genes together, despite their large size, can still create a positional effect. We believe that gene choice, genomic DNA fragment size and the presence of endogenous insulator elements are critical variables.


Assuntos
Cromossomos Artificiais Bacterianos/genética , Genes Reporter , Mutagênese Insercional/métodos , Animais , Clonagem Molecular , DNA/genética , Dosagem de Genes , Técnicas de Transferência de Genes , Vetores Genéticos , Camundongos , Camundongos Transgênicos , Recombinação Genética
17.
Biochem Biophys Res Commun ; 376(2): 375-9, 2008 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-18789892

RESUMO

To effectively treat osteoporosis and other bone-loss disorders, small compounds that potently induce bone formation are needed. The present study initially attempted to establish a monitoring system that could detect osteogenic differentiation easily, precisely, and noninvasively. For this purpose, we established pre-osteoblastic MC3T3E1 cells stably transfected with the GFP reporter gene driven by a 2.3 kb fragment of rat type I collagen promoter (Col1a1GFP-MC3T3E1). Among these cells, we selected a clone that fluoresced upon osteogenic stimulation by BMP2. The GFP fluorescence intensity corresponded well to the intensity of alkaline phosphatase (ALP) staining and to the level of osteocalcin (Oc) mRNA. Using this system, we screened natural and synthetic compound libraries and thus identified an isoflavone derivative, glabrisoflavone (GI). GI induced ALP staining and Oc mRNA in a dose-dependent manner. The Col1a1GFP-MC3T3E1 system may be useful for identifying novel osteogenic drugs.


Assuntos
Técnicas Biossensoriais/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Fosfatase Alcalina/análise , Fosfatase Alcalina/biossíntese , Animais , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/farmacologia , Linhagem Celular , Colágeno Tipo I/genética , Fluorescência , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Isoflavonas/farmacologia , Camundongos , Osteocalcina/análise , Osteocalcina/biossíntese , Ratos , Fator de Crescimento Transformador beta/farmacologia
18.
Dev Biol ; 316(2): 458-70, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18280462

RESUMO

Our laboratory and others have shown that overexpression of Dlx5 stimulates osteoblast differentiation. Dlx5(-/-)/Dlx6(-/-) mice have more severe craniofacial and limb defects than Dlx5(-/-), some of which are potentially due to defects in osteoblast maturation. We wished to investigate the degree to which other Dlx genes compensate for the lack of Dlx5, thus allowing normal development of the majority of skeletal elements in Dlx5(-/-) mice. Dlx gene expression in cells from different stages of the osteoblast lineage isolated by FACS sorting showed that Dlx2, Dlx5 and Dlx6 are expressed most strongly in less mature osteoblasts, whereas Dlx3 is very highly expressed in differentiated osteoblasts and osteocytes. In situ hybridization and Northern blot analysis demonstrated the presence of endogenous Dlx3 mRNA within osteoblasts and osteocytes. Dlx3 strongly upregulates osteoblastic markers with a potency comparable to Dlx5. Cloned chick or mouse Dlx6 showed stimulatory effects on osteoblast differentiation. Our results suggest that Dlx2 and Dlx6 have the potential to stimulate osteoblastic differentiation and may compensate for the absence of Dlx5 to produce relatively normal osteoblastic differentiation in Dlx5 knockout mice, while Dlx3 may play a distinct role in late stage osteoblast differentiation and osteocyte function.


Assuntos
Proteínas de Homeodomínio/genética , Osteoblastos/fisiologia , Osteócitos/fisiologia , Fatores de Transcrição/genética , Animais , Animais Recém-Nascidos , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Diferenciação Celular , Clonagem Molecular , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Osteoblastos/citologia , Osteócitos/citologia , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Células Estromais/citologia , Células Estromais/fisiologia
19.
Dev Growth Differ ; 49(6): 515-21, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17555518

RESUMO

The homeodomain transcription factor Dlx5 has been implicated in the regulation of chondrocyte and osteoblast differentiation during endochondral ossification in the developing limb. In a gain-of-function approach to directly investigate the role of Dlx5 in chondrocyte maturation, we have used cartilage-specific Col2a1-Dlx5 promoter/enhancer constructs to target overexpression of Dlx5 to the differentiating cartilage models of the limbs of transgenic mice. Targeted overexpression of Dlx5 in cartilage rudiments results in the formation of shortened skeletal elements containing excessive numbers of hypertrophic chondrocytes and expanded domains of expression of Ihh and type X collagen, molecular markers of hypertrophic maturation. This suggests that hypertrophic differentiation is enhanced in response to Dlx5 misexpression. Skeletal elements overexpressing Dlx5 also exhibit a marked reduction in the zone of proliferation, indicating that overexpression of Dlx5 reduces chondrocyte proliferation concomitant with promoting hypertrophic maturation. Taken together these results indicate that Dlx5 is a positive regulator of chondrocyte maturation during endochondral ossification, and suggest that it regulates the process at least in part by promoting the conversion of immature proliferating chondrocytes into hypertrophying chondrocytes; a critical step in the maturation process.


Assuntos
Desenvolvimento Ósseo/fisiologia , Diferenciação Celular/fisiologia , Condrócitos/citologia , Extremidades/embriologia , Proteínas de Homeodomínio/fisiologia , Animais , Desenvolvimento Ósseo/genética , Diferenciação Celular/genética , Condrócitos/patologia , Colágeno Tipo II/genética , Extremidades/anatomia & histologia , Proteínas de Homeodomínio/genética , Hipertrofia , Deformidades Congênitas dos Membros/embriologia , Deformidades Congênitas dos Membros/genética , Deformidades Congênitas dos Membros/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
20.
FASEB J ; 21(8): 1777-87, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17317722

RESUMO

To identify potent bioactive factors for in vivo tissue regeneration by comprehensive screening remains a challenge for regenerative medicine. Here we report the development of an ES cell-based monitoring system for osteogenic differentiation, the identification of a potent combination of osteogenic genes using such a system, and an evaluation of its therapeutic potentials. ES cells were isolated from mice carrying a transgene expressing GFP driven by the 2.3 kb fragment of rat type I collagen alpha1 promoter. Using these cells engineered to fluoresce on osteogenic differentiation, we screened cDNA libraries and combinations of major osteogenesis-related genes. Among them, the combination of constitutively active activin receptor-like kinase 6 (caALK6) and runt-related transcription factor 2 (Runx2) was the minimal unit that induced fluorescence. The combination efficiently induced osteogenic differentiation in various cell types, including terminally differentiated nonosteogenic cells. The cooperative action of the combination occurred through protein stabilization of core binding factor beta (Cbfb), induction of Runx2-Cbfb complex formation, and its DNA binding. Furthermore, transplantation of a monolayer sheet of fibroblasts transduced with the combination achieved bone regeneration within 4 wk in mouse calvarial bone defects. Thus, we successfully identified the potent combination of genes for bone regeneration, which helped broaden cell sources.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/fisiologia , Regeneração Óssea/genética , Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Células-Tronco Embrionárias/citologia , Osteogênese/genética , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Diferenciação Celular , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Proteínas de Fluorescência Verde/genética , Camundongos , Ratos , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...