Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Biomater Transl ; 3(1): 3-16, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35837340

RESUMO

Based on studies over the last several decades, the self-renewing skeletal lineages derived from bone marrow stroma could be an ideal source for skeletal tissue engineering. However, the markers for osteogenic precursors; i.e., bone marrowderived skeletal stem cells (SSCs), in association with other cells of the marrow stroma (bone marrow stromal cells, BMSCs) and their heterogeneous nature both in vivo and in vitro remain to be clarified. This review aims to highlight: i) the importance of distinguishing BMSCs/SSCs from other "mesenchymal stem/stromal cells", and ii) factors that are responsible for their heterogeneity, and how these factors impact on the differentiation potential of SSCs towards bone. The prospective role of SSC enrichment, their expansion and its impact on SSC phenotype is explored. Emphasis has also been given to emerging single cell RNA sequencing approaches in scrutinizing the unique population of SSCs within the BMSC population, along with their committed progeny. Understanding the factors involved in heterogeneity may help researchers to improvise their strategies to isolate, characterize and adopt best culture practices and source identification to develop standard operating protocols for developing reproducible stem cells grafts. However, more scientific understanding of the molecular basis of heterogeneity is warranted that may be obtained from the robust high-throughput functional transcriptomics of single cells or clonal populations.

2.
Nat Cell Biol ; 21(7): 801-811, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31209293

RESUMO

Despite many reports of putative stem-cell-based treatments in genetic and degenerative disorders or severe injuries, the number of proven stem cell therapies has remained small. In this Review, we survey advances in stem cell research and describe the cell types that are currently being used in the clinic or are close to clinical trials. Finally, we analyse the scientific rationale, experimental approaches, caveats and results underpinning the clinical use of such stem cells.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Células-Tronco Embrionárias/transplante , Terapia Genética , Regeneração/genética , Células da Medula Óssea/metabolismo , Terapia Baseada em Transplante de Células e Tecidos/métodos , Terapia Genética/métodos , Humanos , Regeneração/fisiologia , Pesquisa com Células-Tronco
4.
Methods Mol Biol ; 1130: 279-293, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24482181

RESUMO

Populations of bone marrow stromal cells (BMSCs, also known as bone marrow-derived "mesenchymal stem cells") contain a subset of cells that are able to recapitulate the formation of a bone/marrow organ (skeletal stem cells, SSCs). The biological properties of BMSC cultures are assessed by a variety of assays, both in vitro and in vivo. Application of these assays in an appropriate fashion provides a great deal of information on the role of BMSCs, and the subset of SSCs, in health and in disease.


Assuntos
Ensaio de Unidades Formadoras de Colônias , Células-Tronco Mesenquimais/fisiologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Camundongos
5.
J Tissue Eng Regen Med ; 7(3): 226-35, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22052864

RESUMO

Autologous transplantation of human bone marrow stromal cells (BMSCs) has been successfully used for bone reconstruction. However, in order to advance this approach into the mainstream of bone tissue engineering, the conditions for BMSC cultivation and transplantation must be optimized. In a recent report, cultivation with dexamethasone (Dex) significantly increased bone formation by human BMSCs in vivo. Based on this important conclusion, we analysed the data accumulated by our laboratory, where human BMSCs have been routinely generated using media both with and without a combination of two osteogenic supplements: Dex at 10(-8) m and ascorbic acid phosphate (AscP) at 10(-4) m. Our data demonstrate that for 22/24 donors, BMSC strains propagated with and without Dex/AscP formed similar amounts of bone in vivo. Thus, human BMSCs do not appear to need to be induced to osteogenic differentiation ex vivo prior to transplantation. Similarly, for 12/14 donors, BMSC strains cultured with and without Dex/AscP formed haematopoietic territories to a comparable extent. While Dex/AscP did not increase bone formation, they significantly stimulated BMSC in vitro proliferation without affecting the number of BMSC colonies formed by the colony-forming units-fibroblasts. We conclude that for the substantial majority of donors, Dex/AscP have no effect on the ability of BMSCs to form bone and myelosupportive stroma in vivo. However, due to increased BMSC proliferation, the total osteogenic population obtained from a single marrow sample is larger after cultivation with Dex/AscP than without them. Secondary to increased BMSC proliferation, Dex/AscP may stimulate bone formation if BMSCs and/or the transplantation system are less than optimal. Published 2011. This article is a U.S. Government work and is in the public domain in the USA.


Assuntos
Meios de Cultura/farmacologia , Hematopoese/efeitos dos fármacos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Osteogênese/efeitos dos fármacos , Adolescente , Adulto , Idoso , Animais , Ácido Ascórbico/farmacologia , Proliferação de Células/efeitos dos fármacos , Criança , Dexametasona/farmacologia , Feminino , Humanos , Hospedeiro Imunocomprometido/efeitos dos fármacos , Lactente , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Pessoa de Meia-Idade , Adulto Jovem
6.
Tissue Eng Part B Rev ; 17(6): 423-30, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21797663

RESUMO

Based on the extensive investigation of various ways to regenerate bone, bone marrow stromal cells, in conjunction with ceramic scaffolds, show great promise for application in human patients, and are already in use in a limited number of clinical trials. In preparing for clinical trials, scale-up current good manufacturing processes (cGMP) must incorporate the use of appropriate assays to ensure that the resulting cell product has maintained its biological activity. Future developments are needed to identify better scaffolds, and better ways to deliver cells with either injectable carriers, or by developing techniques to aide in their escape from the circulation and their incorporation into the pre-existing tissue. Lastly, development of methods that faithfully direct pluripotent stem cell differentiation into populations of osteogenic precursors (and ideally, containing skeletal stem cells) represents a new challenge in the field of bone regeneration, but also offer new opportunities to not only to study the biology of bone formation, but also to develop a robust cell source for bone regeneration.


Assuntos
Células da Medula Óssea/citologia , Regeneração Óssea/fisiologia , Separação Celular , Transplante de Células , Humanos , Células Estromais/citologia , Células Estromais/transplante
7.
Stem Cells Dev ; 20(2): 269-87, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20590404

RESUMO

The derivation of osteogenic cells from human embryonic stem cells (hESCs) or from induced pluripotent stem cells for bone regeneration would be a welcome alternative to the use of adult stem cells. In an attempt to promote hESC osteogenic differentiation, cells of the HSF-6 line were cultured in differentiating conditions in vitro for prolonged periods of time ranging from 7 to 14.5 weeks, followed by in vivo transplantation into immunocompromised mice in conjunction with hydroxyapatite/tricalcium phosphate ceramic powder. Twelve different medium compositions were tested, along with a number of other variables in culture parameters. In differentiating conditions, HSF-6-derived cells demonstrated an array of diverse phenotypes reminiscent of multiple tissues, but after a few passages, acquired a more uniform, fibroblast-like morphology. Eight to 16 weeks post-transplantation, a group of transplants revealed the formation of histologically proven bone of human origin, including broad areas of multiple intertwining trabeculae, which represents by far the most extensive in vivo bone formation by the hESC-derived cells described to date. Knockout-Dulbecco's modified Eagle's medium-based media with fetal bovine serum, dexamethasone, and ascorbate promoted more frequent bone formation, while media based on α-modified minimum essential medium promoted teratoma formation in 12- to 20-week-old transplants. Transcription levels of pluripotency-related (octamer binding protein 4, Nanog), osteogenesis-related (collagen type I, Runx2, alkaline phosphatase, and bone sialoprotein), and chondrogenesis-related (collagen types II and X, and aggrecan) genes were not predictive of either bone or teratoma formation. The most extensive bone was formed by the strains that, following 4 passages in monolayer conditions, were cultured for 23 to 25 extra days on the surface of hydroxyapatite/tricalcium phosphate particles, suggesting that coculturing of hESC-derived cells with osteoconductive material may increase their osteogenic potential. While none of the conditions tested in this study, and elsewhere, ensured consistent bone formation by hESC-derived cells, our results may elucidate further directions toward the construction of bone on the basis of hESCs or an individual's own induced pluripotent stem cells.


Assuntos
Células-Tronco Embrionárias/citologia , Osteogênese , Animais , Diferenciação Celular , Linhagem Celular , Células-Tronco Embrionárias/fisiologia , Feminino , Perfilação da Expressão Gênica , Humanos , Hospedeiro Imunocomprometido , Cariotipagem , Camundongos , Camundongos Nus , Osteoblastos/citologia , Gravidez , Transplante de Células-Tronco , Teratoma/etiologia
8.
J Mol Endocrinol ; 45(6): 355-64, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20841428

RESUMO

Activating mutations of the GNAS gene, which causes fibrous dysplasia of bone (FD), lead to remarkable changes in the properties of skeletal progenitors, and it is these changes that mediate the pathological effect of this gene on bone. Mutated skeletal stem cells lose the ability to differentiate into adipocytes, and to maintain in situ, and transfer heterotopically, the hematopoietic microenvironment, leading to abnormal bone marrow histology in FD. They overexpress molecular effectors of osteoclastogenesis, thus promoting inappropriate bone resorption leading to fragility of FD bone. They express the phosphate-regulating hormone FGF-23 at normal levels, whose excess in the serum of FD patients correlates with the mass of osteogenic cells within FD lesions, leading to osteomalacia and deformity of the FD bone, and revealing that bone is an endocrine organ regulating renal handling of phosphate. Mechanisms of allelic selection and stem cell selection occur in mutated skeletal stem cells and contribute to the inherent diversity and evolution over time in FD. The definition of the etiological role of GNAS mutations marks the watershed between many decades of descriptive observation and the definition of cellular and molecular mechanisms that would explain and hopefully allow for a cure for the disease. Placing stem cells at center stage has permitted substantial advances in one decade, and promises more for the one to come.


Assuntos
Osso e Ossos/citologia , Osso e Ossos/metabolismo , Displasia Fibrosa Óssea/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Células-Tronco/metabolismo , Animais , Fator de Crescimento de Fibroblastos 23 , Displasia Fibrosa Óssea/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Humanos , Modelos Biológicos , Mutação , Células-Tronco/citologia
9.
Hum Gene Ther ; 21(9): 1057-66, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20649485

RESUMO

At the turn of a decade of intensive wishful thinking, "mesenchymal stem cells" are changing their profile, while retaining their charm. As hopes to turn bone into brain or vice versa seem on the wane, we learn (1) that the archetypal "mesenchymal stem cell," the skeletal stem cell found in the bone marrow, can be directly identified as a specialized type of mural cell/pericyte, found in the wall of sinusoids and long known as adventitial reticular cells; (2) that bone marrow skeletal stem cells are also defined by expression of CD146, and can self-renew in vivo, while giving rise to skeletal tissues, and therefore earn consideration as bona fide stem cells; (3) that a broader class of microvascular mural cells endowed with clonogenicity and progenitor properties may exist in other tissues, although their true potency needs to be firmly established by stringent assays and thorough comparisons across tissues; (4) that bone marrow skeletal stem cells display unique angiopoietic and hematopoietic niche-related functions, consisting in their ability to transfer the hematopoietic microenvironment and to guide the assembly of microvascular networks, which seem to define their inherent biology; and (5) that use of skeletal stem cells as disease models, and as models of high-risk strategies for cell and gene therapy specifically in incurable skeletal diseases, may provide new challenges for the next decade, and perhaps reward for medicine in the one that follows.


Assuntos
Doenças Ósseas/terapia , Células da Medula Óssea/citologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Proliferação de Células , Ensaio de Unidades Formadoras de Colônias , Humanos , Pericitos/citologia , Engenharia Tecidual
10.
Expert Rev Mol Diagn ; 10(4): 465-80, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20465501

RESUMO

Multiple myeloma (MM) is the second most common hematological malignancy, with an overall survival of 4-6 years. It is always preceded by a premalignant stage called monoclonal gammopathy of unknown significance (MGUS). Importantly, at this time we lack reliable predictors to determine who will progress from MGUS to MM, and who will remain stable. The bone marrow microenvironment plays a key role in myelomagenesis (growth, survival and migration of malignant plasma cells). In the present review, we summarize and discuss our current understanding of the bone marrow microenvironment and its compartments in relation to myelomagenesis. Although it remains to be proven, we believe that an improved characterization of the cellular constituents, the extracellular matrix components and the soluble factors of the bone marrow could open up novel avenues to better understand underlying mechanisms of the transformation from MGUS to MM. Ultimately, this will lead to the development of early treatment of high-risk precursor disease aimed to delay/prevent MM.


Assuntos
Medula Óssea , Meio Ambiente , Mieloma Múltiplo , Biomarcadores/metabolismo , Medula Óssea/metabolismo , Medula Óssea/patologia , Células da Medula Óssea/metabolismo , Progressão da Doença , Diagnóstico Precoce , Humanos , Gamopatia Monoclonal de Significância Indeterminada/fisiopatologia , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Prognóstico , Fatores de Risco
11.
J Bone Miner Res ; 25(5): 1103-16, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19874199

RESUMO

Human skeletal progenitors were engineered to stably express R201C mutated, constitutively active Gs alpha using lentiviral vectors. Long-term transduced skeletal progenitors were characterized by an enhanced production of cAMP, indicating the transfer of the fundamental cellular phenotype caused by activating mutations of Gs alpha. Like skeletal progenitors isolated from natural fibrous dysplasia (FD) lesions, transduced cells could generate bone but not adipocytes or the hematopoietic microenvironment on in vivo transplantation. In vitro osteogenic differentiation was noted for the lack of mineral deposition, a blunted upregulation of osteocalcin, and enhanced upregulation of other osteogenic markers such as alkaline phosphatase (ALP) and bone sialoprotein (BSP) compared with controls. A very potent upregulation of RANKL expression was observed, which correlates with the pronounced osteoclastogenesis observed in FD lesions in vivo. Stable transduction resulted in a marked upregulation of selected phosphodiesterase (PDE) isoform mRNAs and a prominent increase in total PDE activity. This predicts an adaptive response in skeletal progenitors transduced with constitutively active, mutated Gs alpha. Indeed, like measurable cAMP levels, the differentiative responses of transduced skeletal progenitors were profoundly affected by inhibition of PDEs or lack thereof. Finally, using lentiviral vectors encoding short hairpin (sh) RNA interfering sequences, we demonstrated that selective silencing of the mutated allele is both feasible and effective in reverting the aberrant cAMP production brought about by the constitutively active Gs alpha and some of its effects on in vitro differentiation of skeletal progenitors.


Assuntos
Células da Medula Óssea/metabolismo , Displasia Fibrosa Óssea/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Células-Tronco/fisiologia , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Displasia Fibrosa Óssea/patologia , Displasia Fibrosa Óssea/fisiopatologia , Vetores Genéticos , Humanos , Lentivirus/genética , Osteogênese/genética , Fenótipo , Diester Fosfórico Hidrolases/metabolismo , Células Estromais/metabolismo , Transdução Genética
12.
Tissue Eng Part A ; 14(12): 1949-58, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18800877

RESUMO

BACKGROUND: The in vivo transplantation assay has become a valuable tool for assessing the osteogenic potential of diverse cell populations. It has required that cells are cotransplanted with a matrix into recipient animals using large incisions and extensive dissections. Here, we demonstrate that transplants of an osteogenic cell population, bone marrow stromal cells (BMSCs), are capable of assembling into mature bone organs when injected as suspensions of cells and a particulate matrix. METHODS: Human BMSCs, along with hydroxyapatite/tricalcium phosphate (HA/TCP) particles, were placed either into the dorsal subcutaneous space or onto the calvarium of immunodeficient mice, either via injection or via a wide operative exposure. Transplants were harvested from 7 to 110 weeks later; their histologic and mechanical properties and their cellular origin were analyzed. RESULTS: A total of 43 transplants were evaluated. The extent of new bone and hematopoiesis, the bone's adherence to the underlying mouse calvarium, and the bone elastic modulus and hardness were comparable between the two groups. In situ hybridization confirmed a human origin of the new bone. CONCLUSIONS: Our data indicate that BMSCs and HA/TCP particles, when injected as a suspension, can assemble into mature bone organs, and that this bone has histologic and mechanical properties similar to bone formed in standard transplants delivered through a large incision. These results open the possibility for assessing the osteogenic capacities of cell populations, for modeling bone formation and repair and for treating bone deficits, all in the context of minimal surgical intervention or soft tissue disruption.


Assuntos
Células da Medula Óssea/citologia , Fosfatos de Cálcio/administração & dosagem , Durapatita/administração & dosagem , Osteogênese/fisiologia , Células Estromais/transplante , Administração Cutânea , Adolescente , Animais , Fenômenos Biomecânicos , Osso e Ossos/citologia , Feminino , Humanos , Injeções , Masculino , Camundongos , Crânio/citologia , Células Estromais/citologia , Suspensões
13.
J Bone Miner Res ; 23(11): 1731-40, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18597624

RESUMO

We studied the role of somatic mosaicism in fibrous dysplasia of bone (FD) within the context of skeletal ("mesenchymal") stem cells by assessing the frequency of mutated colony forming unit-fibroblasts (CFU-Fs) from FD lesions, and in some cases, from unaffected sites, in a series of patients. There was a tight inverse correlation between the percentage mutant CFU-F versus age, suggesting demise of mutant stem cells caused by exuberant apoptosis noted in samples from young patients. In older patients, either partially or completely normal bone/marrow histology was observed. On in vivo transplantation, FD ossicles were generated only by cell strains in which mutant CFU-Fs were identified. Strains that lacked mutant CFU-F (but were mutation positive) failed to regenerate an FD ossicle. These data indicate that GNAS mutations are only pathogenic when in clonogenic skeletal stem cells. From these data, we have evolved the novel concept of "normalization" of FD. As a lesion ages, mutant stem cells fail to self-renew, and their progeny are consumed by apoptosis, whereas residual normal stem cells survive, self-renew, and enable formation of a normal structure. This suggests that activating GNAS mutations disrupt a pathway that is required for skeletal stem cell self-renewal.


Assuntos
Envelhecimento/patologia , Displasia Fibrosa Óssea/genética , Displasia Fibrosa Óssea/patologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Músculo Esquelético/citologia , Mutação/genética , Células-Tronco/patologia , Adolescente , Adulto , Animais , Apoptose , Células da Medula Óssea/patologia , Células Cultivadas , Criança , Cromograninas , Células Clonais , Ensaio de Unidades Formadoras de Colônias , Análise Mutacional de DNA , Feminino , Displasia Fibrosa Óssea/diagnóstico por imagem , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Ácidos Nucleicos Peptídicos/metabolismo , Cintilografia , Células Estromais/patologia , Células Estromais/transplante
14.
Cell Stem Cell ; 2(4): 313-9, 2008 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-18397751

RESUMO

The concept of mesenchymal stem cells has gained wide popularity. Despite the rapid growth of the field, uncertainties remain with respect to the defining characteristics of these cells, including their potency and self-renewal. These uncertainties are reflected in a growing tendency to question the very use of the term. This commentary revisits the experimental origin of the concept of the population(s) referred to as mesenchymal stem cells and the experimental framework required to assess their stemness and function.


Assuntos
Células-Tronco Mesenquimais/citologia , Biologia Celular/história , Diferenciação Celular/fisiologia , Proliferação de Células , História do Século XX , História do Século XXI , Humanos
15.
J Cell Biochem ; 104(5): 1865-78, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18404666

RESUMO

Wnt-induced secreted protein-1 (WISP-1), like other members of the CCN family, is expressed in skeletal tissues. Its mechanism of action remains unknown. Expression of WISP-1 was analyzed in human bone marrow stroma cells (hBMSC) by RT-PCR. We identified two major transcripts corresponding to those of full-length WISP-1, and of the splice variant WISP-1va which lacks a putative BMP/TGF-beta binding site. To investigate the function of WISP-1 in bone, hBMSC cultures were treated with recombinant human (rh)WISP-1 and analyzed for proliferation and osteogenic differentiation. WISP-1 treatment increased both BrdU incorporation and alkaline phosphatase (AP) activity. Considering the known functional synergy found between the TGF-beta super-family and members of the CCN family, we next tested the effect of WISP-1 on TGF-beta1 activity. We found that rhWISP-1 could reduce rhTGF-beta1 induced BrdU incorporation. Similarly, rhTGF-beta1 inhibited rhWISP-1 induction of AP activity. To explore functional differences between the WISP-1 variants, WISP-1 or WISP-1va were transfected into hBMSC. Both variants could strongly induce BrdU incorporation. However, there were no effects of either variant on AP activity without an additional osteogenic stimulus such as TGF-beta1. Taken together our results suggest a functional relationship between WISP-1 and TGF-beta1. To further define this relationship we analyzed the effect of WISP-1 on TGF-beta signaling. rhWISP-1 significantly reduced TGF-beta1 induced phosphorylation of Smad-2. Our data indicates that full-length WISP-1 and its variant WISP-1va are modulators of proliferation and osteogenic differentiation, and may be novel regulators of TGF-beta1 signaling in osteoblast-like cells.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Osteoblastos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Processamento Alternativo/efeitos dos fármacos , Sequência de Aminoácidos , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Proteínas de Sinalização Intercelular CCN , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Dados de Sequência Molecular , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/farmacologia , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
16.
Methods Mol Med ; 140: 83-99, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18085204

RESUMO

Skeletal stem cells (SSCs, commonly referred to as "mesenchymal" stem cells) are found in the bone marrow stromal cell (BMSC) fraction of post-natal bone marrow. They can be isolated in culture as adherent, clonogenic cells endowed with the ability to grow and differentiate into multiple lineages, all of which correspond to tissues that are integral parts of the skeleton. The multipotency of SSCs is probed by in vivo transplantation assays. The ability of SSCs to generate a cell strain competent to form significant amounts of bone in vivo has led to the formulation of preclinical models of bone repair.


Assuntos
Células da Medula Óssea , Células-Tronco Mesenquimais , Engenharia Tecidual/métodos , Animais , Materiais Biocompatíveis/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Fosfatos de Cálcio/metabolismo , Técnicas de Cultura de Células , Células Cultivadas , Colágeno/metabolismo , Humanos , Hidroxiapatitas/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Transplante de Células-Tronco
17.
Pediatr Endocrinol Rev ; 4 Suppl 4: 386-94, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17982385

RESUMO

Stem cells have become a major area of interest in the treatment of human disease, but more recently, stem cells have come to be appreciated as the cause of disease. Fibrous dysplasia of bone and the McCune-Albright Syndrome evolve from activating missense mutations in Gsalpha in pluripotent embryonic stem cells. The legacy of these mutations remains in a population of mutated multipotent post-natal skeletal stem cells ("mesenchymal" stem cells), which direct the formation of abnormal bone and a fibrotic marrow in fibrous dysplasia. Future therapeutic approaches for the treatment of fibrous dysplasia, the most significant component of the McCune-Albright Syndrome, will depend on a greater understanding of post-natal skeletal stem cell biology and how skeletal stem cells can be manipulated for efficient bone regeneration.


Assuntos
Displasia Fibrosa Óssea/patologia , Displasia Fibrosa Poliostótica/patologia , Células-Tronco Mesenquimais , Células-Tronco Pluripotentes/patologia , Humanos
18.
Pediatr Endocrinol Rev ; 4 Suppl 4: 401-11, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17982387

RESUMO

Fibrous dysplasia (FD) is the most serious and least understood clinical expression in patients with activating mutations of the GNAS gene. Since the discovery of the causative mutation, important progress has been made in the understanding of the pathology of FD and the pathogenesis of bone lesions. The histology of FD has been reinterpreted in light of the pathological effect of the genetic lesions on mutated skeletal stem cells. True histological hallmarks of the disease have emerged, along with genetic analysis, as additional tools to establish the correct diagnosis. Furthermore, the recognition of FD as a disease of excess, abnormal and imperfect bone formation has helped to explain relevant mechanisms of its clinical morbidity, based on which potential specific therapeutic approaches may be developed in the near future.


Assuntos
Calcificação Fisiológica , Displasia Fibrosa Óssea/patologia , Displasia Fibrosa Poliostótica/patologia , Displasia Fibrosa Óssea/diagnóstico por imagem , Displasia Fibrosa Poliostótica/diagnóstico por imagem , Humanos , Radiografia
19.
Exp Hematol ; 35(6): 995-1004, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17960668

RESUMO

OBJECTIVE: Bone marrow stromal cells (BMSCs) include multipotent cells with the ability to form mature bone organs upon in vivo transplantation. Hematopoiesis in these bone organs has been ascribed to the action of skeletal stem cells, which are capable of differentiating towards bone and hematopoiesis-supporting stroma. Yet, the creation of hematopoietic territories may be in part a natural consequence of the formation of a sufficiently mature and large bone microenvironment. Here, we describe, for the first time, a relationship between BMSC numbers and the extent of bone/hematopoiesis formation in heterotopic transplants. METHODS: Human BMSCs were transplanted along with hydroxyapatite/tricalcium phosphate, utilizing a spectrum of dosages, into immunotolerant mice; the transplants were followed for up to 29 months. RESULTS: The extent of bone and hematopoiesis formation increased with increasing BMSC numbers; however, the relationship was sigmoid in character, and a threshold number of BMSCs was necessary for extensive bone formation or any hematopoiesis. Hematopoiesis only occurred in conjunction with extensive bone formation, and no hematopoiesis occurred where bone formation was poor. Consistent with our earlier studies of long-term BMSC transplantation, the transplants underwent a change in bone morphology but not bone content after 8 weeks. CONCLUSION: Our results have provided evidence that the formation of both hematopoiesis and a mature bone organ is as much a consequence of a sufficiently high local density of bone marrow stromal cells as it is the product of skeletal stem cell action.


Assuntos
Células da Medula Óssea , Hematopoese , Osteogênese , Células Estromais , Transplante Heterotópico , Adolescente , Animais , Contagem de Células , Diferenciação Celular , Transplante de Células , Feminino , Humanos , Hospedeiro Imunocomprometido , Masculino , Camundongos , Células-Tronco/citologia , Células Estromais/citologia , Células Estromais/transplante , Engenharia Tecidual
20.
Cell ; 131(2): 324-36, 2007 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-17956733

RESUMO

The identity of cells that establish the hematopoietic microenvironment (HME) in human bone marrow (BM), and of clonogenic skeletal progenitors found in BM stroma, has long remained elusive. We show that MCAM/CD146-expressing, subendothelial cells in human BM stroma are capable of transferring, upon transplantation, the HME to heterotopic sites, coincident with the establishment of identical subendothelial cells within a miniature bone organ. Establishment of subendothelial stromal cells in developing heterotopic BM in vivo occurs via specific, dynamic interactions with developing sinusoids. Subendothelial stromal cells residing on the sinusoidal wall are major producers of Angiopoietin-1 (a pivotal molecule of the HSC "niche" involved in vascular remodeling). Our data reveal the functional relationships between establishment of the HME in vivo, establishment of skeletal progenitors in BM sinusoids, and angiogenesis.


Assuntos
Medula Óssea/irrigação sanguínea , Osso e Ossos/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Mesenquimais/citologia , Angiopoietina-1/metabolismo , Animais , Osso e Ossos/metabolismo , Antígeno CD146/metabolismo , Diferenciação Celular , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Células Estromais/citologia , Células Estromais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA