Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Sci Rep ; 8(1): 15618, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30353078

RESUMO

The vascular wall within adipose tissue is a source of mesenchymal progenitors, referred to as perivascular stem/stromal cells (PSC). PSC are isolated via fluorescence activated cell sorting (FACS), and defined as a bipartite population of pericytes and adventitial progenitor cells (APCs). Those factors that promote the differentiation of PSC into bone or fat cell types are not well understood. Here, we observed high expression of WISP-1 among human PSC in vivo, after purification, and upon transplantation in a bone defect. Next, modulation of WISP-1 expression was performed, using WISP-1 overexpression, WISP-1 protein, or WISP-1 siRNA. Results demonstrated that WISP-1 is expressed in the perivascular niche, and high expression is maintained after purification of PSC, and upon transplantation in a bone microenvironment. In vitro studies demonstrate that WISP-1 has pro-osteogenic/anti-adipocytic effects in human PSC, and that regulation of BMP signaling activity may underlie these effects. In summary, our results demonstrate the importance of the matricellular protein WISP-1 in regulation of the differentiation of human stem cell types within the perivascular niche. WISP-1 signaling upregulation may be of future benefit in cell therapy mediated bone tissue engineering, for the healing of bone defects or other orthopaedic applications.


Assuntos
Osso e Ossos/metabolismo , Osso e Ossos/fisiologia , Proteínas de Sinalização Intercelular CCN/metabolismo , Gorduras/metabolismo , Osteogênese/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Tecido Adiposo/metabolismo , Tecido Adiposo/fisiologia , Diferenciação Celular/fisiologia , Separação Celular/métodos , Células Cultivadas , Microambiente Celular/fisiologia , Citometria de Fluxo/métodos , Humanos , Pericitos/metabolismo , Pericitos/fisiologia , Engenharia Tecidual/métodos , Regulação para Cima/fisiologia
2.
Crit Rev Oncog ; 23(5-6): 333-346, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30311564

RESUMO

Since the discovery of autophagy in the mid-2000s, the interest in autophagy-related processes within the scientific community has been burgeoning. Countless authors have investigated its function in cellular homeostasis, but arguably of higher importance is its role during pathology. Although primarily a catabolic process, in cancer cells autophagy has numerous downstream effects, being observed to be both pro- and anti-apoptotic. One of the primary factors mediating this differential role of autophagy is the accumulation or sequestration of reactive oxygen species (ROS). Until recently, despite its increasing popularity in the Western world, the efficacy of herbal supplements has been largely anecdotal. Herein, we reviewed the ten most commonly studied herbs in cancer research and their impact on ROS regulation, on the activation and inhibition of autophagy, and ultimately on cancer cell death.


Assuntos
Autofagia/efeitos dos fármacos , Medicina Herbária , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Biomarcadores , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Medicina Herbária/métodos , Humanos , Neoplasias/etiologia , Neoplasias/patologia , Oxirredução/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
4.
J Bone Miner Res ; 33(10): 1813-1825, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29905970

RESUMO

Contactin-associated protein-like 4 (Cntnap4) is a member of the neurexin superfamily of transmembrane molecules that have critical functions in neuronal cell communication. Cntnap4 knockout mice display decreased presynaptic gamma-aminobutyric acid (GABA) and increased dopamine release that is associated with severe, highly penetrant, repetitive, and perseverative movements commonly found in human autism spectrum disorder patients. However, no known function of Cntnap4 has been revealed besides the nervous system. Meanwhile, secretory protein neural EGFL-like 1 (Nell-1) is known to exert potent osteogenic effects in multiple small and large animal models without the off-target effects commonly found with bone morphogenetic protein 2. In this study, while searching for a Nell-1-specific cell surface receptor during osteogenesis, we identified and validated a ligand/receptor-like interaction between Nell-1 and Cntnap4 by demonstrating: 1) Nell-1 and Cntnap4 colocalization on the surface of osteogenic-committed cells; 2) high-affinity interaction between Nell-1 and Cntnap4; 3) abrogation of Nell-1-responsive Wnt and MAPK signaling transduction, as well as osteogenic effects, via Cntnap4 knockdown; and 4) replication of calvarial cleidocranial dysplasias-like defects observed in Nell-1-deficient mice in Wnt1-Cre-mediated Cntnap4-knockout transgenic mice. In aggregate, these findings indicate that Cntnap4 plays a critical role in Nell-1-responsive osteogenesis. Further, this is the first functional annotation for Cntnap4 in the musculoskeletal system. Intriguingly, Nell-1 and Cntnap4 also colocalize on the surface of human hippocampal interneurons, implicating Nell-1 as a potential novel ligand for Cntnap4 in the nervous system. This unexpected characterization of the ligand/receptor-like interaction between Nell-1 and Cntnap4 indicates a novel biological functional axis for Nell-1 and Cntnap4 in osteogenesis and, potentially, in neural development and function. © 2018 American Society for Bone and Mineral Research.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Glicoproteínas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Osteogênese , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Bacteriófago T7/metabolismo , Medula Óssea/metabolismo , Linhagem Celular , Linhagem da Célula , Membrana Celular/metabolismo , Deleção de Genes , Humanos , Integrases/metabolismo , Proteínas de Membrana/química , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Proteínas do Tecido Nervoso/química , Ligação Proteica , Domínios Proteicos , Transdução de Sinais , Crânio/metabolismo
5.
Int J Mol Sci ; 19(1)2018 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-29316655

RESUMO

Neural EGFL like 1 (Nell-1) is essential for chondrogenic differentiation, maturation, and regeneration. Our previous studies have demonstrated that Nell-1's pro-chondrogenic activities are predominantly reliant upon runt-related transcription factor 3 (Runx3)-mediated Indian hedgehog (Ihh) signaling. Here, we identify the nuclear factor of activated T-cells 1 (Nfatc1) as the key transcriptional factor mediating the Nell-1 → Runx3 signal transduction in chondrocytes. Using chromatin immunoprecipitation assay, we were able to determine that Nfatc1 binds to the -833--810 region of the Runx3-promoter in response to Nell-1 treatment. By revealing the Nell-1 → Nfatc1 → Runx3 → Ihh cascade, we demonstrate the involvement of Nfatc1, a nuclear factor of activated T-cells, in chondrogenesis, while providing innovative insights into developing a novel therapeutic strategy for cartilage regeneration and other chondrogenesis-related conditions.


Assuntos
Proteínas de Ligação ao Cálcio/farmacologia , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Glicoproteínas/farmacologia , Fatores de Transcrição NFATC/metabolismo , Regulação para Cima/efeitos dos fármacos , Tecido Adiposo/citologia , Animais , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Condrócitos/citologia , Condrócitos/metabolismo , Condrogênese/efeitos dos fármacos , Subunidade alfa 1 de Fator de Ligação ao Core/deficiência , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/antagonistas & inibidores , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Camundongos , Camundongos Knockout , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/genética , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Tissue Eng Part A ; 24(1-2): 68-80, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28463594

RESUMO

Human perivascular stem/stromal cells (hPSC) are a multipotent mesenchymogenic stromal cell population defined by their perivascular locale. Recent studies have demonstrated the high potential for clinical translation of this fluorescence-activated cell sorting (FACS)-derived cell population for autologous bone tissue engineering. However, the mechanisms underlying the osteogenic differentiation of PSC are incompletely understood. The current study investigates the roles of canonical and noncanonical Wnt signaling in the osteogenic and adipogenic differentiation of PSC. Results showed that both canonical and noncanonical Wnt signaling activity transiently increased during PSC osteogenic differentiation in vitro. Sustained WNT3A treatment significantly decreased PSC osteogenic differentiation. Conversely, sustained treatment with Wnt family member 16 (WNT16), a mixed canonical and noncanonical ligand, increased osteogenic differentiation in a c-Jun N-terminal kinase (JNK) pathway-dependent manner. Conversely, WNT16 knockdown significantly diminished PSC osteogenic differentiation. Finally, WNT16 but not WNT3A increased the adipogenic differentiation of PSC. These results indicate the importance of regulation of canonical and noncanonical Wnt signaling for PSC fate and differentiation. Moreover, these data suggest that WNT16 plays a functional and necessary role in PSC osteogenesis.


Assuntos
Adipogenia/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Proteínas Wnt/farmacologia , Proteína Wnt3A/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Engenharia Tecidual/métodos , Via de Sinalização Wnt/efeitos dos fármacos
7.
Tissue Eng Part A ; 24(5-6): 448-457, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28683667

RESUMO

Human perivascular stem/stromal cells (PSC) are a multipotent mesodermal progenitor cell population defined by their perivascular residence. PSC are most commonly derived from subcutaneous adipose tissue, and recent studies have demonstrated the high potential for clinical translation of this fluorescence-activated cell sorting-derived cell population for bone tissue engineering. Specifically, purified PSC induce greater bone formation than unpurified stroma taken from the same patient sample. In this study, we examined the differences in early innate immune response to human PSC or unpurified stroma (stromal vascular fraction [SVF]) during the in vivo process of bone formation. Briefly, SVF or PSC from the same patient sample were implanted intramuscularly in the hindlimb of severe combined immunodeficient (SCID) mice using an osteoinductive demineralized bone matrix carrier. Histological examination of early inflammatory infiltrates was examined by hematoxylin and eosin and immunohistochemical staining (Ly-6G, F4/80). Results showed significantly greater neutrophilic and macrophage infiltrates within and around SVF in comparison to PSC-laden implants. Differences in early postoperative inflammation among SVF-laden implants were associated with reduced osteogenic differentiation and bone formation. Similar findings were recapitulated with PSC implantation in immunocompetent mice. Exaggerated postoperative inflammation was associated with increased IL-1α, IL-1ß, IFN-γ, and TNF-α gene expression among SVF samples, and conversely increased IL-6 and IL-10 expression among PSC samples. These data document a robust immunomodulatory effect of implanted PSC, and an inverse correlation between host inflammatory cell infiltration and stromal progenitor cell-mediated ossification.


Assuntos
Matriz Óssea , Células Imobilizadas , Imunomodulação , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Osteogênese/imunologia , Animais , Matriz Óssea/imunologia , Matriz Óssea/transplante , Células Imobilizadas/imunologia , Células Imobilizadas/transplante , Citocinas/imunologia , Xenoenxertos , Humanos , Camundongos , Camundongos SCID
8.
PLoS One ; 12(5): e0177308, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28489940

RESUMO

For over 15 years, human subcutaneous adipose tissue has been recognized as a rich source of tissue resident mesenchymal stem/stromal cells (MSC). The isolation of perivascular progenitor cells from human adipose tissue by a cell sorting strategy was first published in 2008. Since this time, the interest in using pericytes and related perivascular stem/stromal cell (PSC) populations for tissue engineering has significantly increased. Here, we describe a set of experiments identifying, isolating and characterizing PSC from canine tissue (N = 12 canine adipose tissue samples). Results showed that the same antibodies used for human PSC identification and isolation are cross-reactive with canine tissue (CD45, CD146, CD34). Like their human correlate, canine PSC demonstrate characteristics of MSC including cell surface marker expression, colony forming unit-fibroblast (CFU-F) inclusion, and osteogenic differentiation potential. As well, canine PSC respond to osteoinductive signals in a similar fashion as do human PSC, such as the secreted differentiation factor NEL-Like Molecule-1 (NELL-1). Nevertheless, important differences exist between human and canine PSC, including differences in baseline osteogenic potential. In summary, canine PSC represent a multipotent mesenchymogenic cell source for future translational efforts in tissue engineering.


Assuntos
Tecido Adiposo/citologia , Separação Celular , Osteogênese , Células Estromais/citologia , Engenharia Tecidual , Animais , Osso e Ossos/citologia , Osso e Ossos/fisiologia , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Separação Celular/métodos , Células Cultivadas , Cães , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes/metabolismo , Células Estromais/metabolismo , Engenharia Tecidual/métodos , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Pharmacol Ther ; 171: 93-103, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27510330

RESUMO

Pericytes and other perivascular stem cells are of growing interest in orthopedics and tissue engineering. Long regarded as simple regulators of angiogenesis and blood pressure, pericytes are now recognized to have MSC (mesenchymal stem cell) characteristics, including multipotentiality, self-renewal, immunoregulatory functions, and diverse roles in tissue repair. Pericytes are typified by characteristic cell surface marker expression (including αSMA, CD146, PDGFRß, NG2, RGS5, among others). Although alone no marker is absolutely specific for pericytes, collectively these markers appear to selectively identify an MSC-like pericyte. The purification of pericytes is most well described as a CD146+CD34-CD45- cell population. Pericytes and other perivascular stem cell populations have been applied in diverse orthopedic applications, including both ectopic and orthotopic models. Application of purified cells has sped calvarial repair, induced spine fusion, and prevented fibrous non-union in rodent models. Pericytes induce these effects via both direct and indirect mechanisms. In terms of their paracrine effects, pericytes are known to produce and secrete high levels of a number of growth and differentiation factors both in vitro and after transplantation. The following review will cover existing studies to date regarding pericyte application for bone and cartilage engineering. In addition, further questions in the field will be pondered, including the phenotypic and functional overlap between pericytes and culture-derived MSC, and the concept of pericytes as efficient producers of differentiation factors to speed tissue repair.


Assuntos
Doenças Musculoesqueléticas/terapia , Pericitos/citologia , Engenharia Tecidual/métodos , Animais , Osso e Ossos/citologia , Cartilagem/citologia , Diferenciação Celular , Humanos , Células-Tronco Mesenquimais/citologia , Doenças Musculoesqueléticas/patologia , Roedores , Células-Tronco/citologia
10.
Hum Pathol ; 58: 24-34, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27498059

RESUMO

Sclerostin (SOST) is an extracellular Wnt signaling antagonist which negatively regulates bone mass. Despite this, the expression and function of SOST in skeletal tumors remain poorly described. Here, we first describe the immunohistochemical staining pattern of SOST across benign and malignant skeletal tumors with bone or cartilage matrix (n=68 primary tumors). Next, relative SOST expression was compared to markers of Wnt signaling activity and osteogenic differentiation across human osteosarcoma (OS) cell lines (n=7 cell lines examined). Results showed immunohistochemical detection of SOST in most bone-forming tumors (90.2%; 46/51) and all cartilage-forming tumors (100%; 17/17). Among OSs, variable intensity and distribution of SOST expression were observed, which highly correlated with the presence and degree of neoplastic bone. Patchy SOST expression was observed in cartilage-forming tumors, which did not distinguish between benign and malignant tumors or correlate with regional morphologic characteristics. Finally, SOST expression varied widely between OS cell lines, with more than 97-fold variation. Among OS cell lines, SOST expression positively correlated with the marker of osteogenic differentiation alkaline phosphatase and did not correlate well with markers of Wnt/ß-catenin signaling activity. In summary, SOST is frequently expressed in skeletal bone- and cartilage-forming tumors. The strong spatial correlation with bone formation and the in vitro expression patterns are in line with the known functions of SOST in nonneoplastic bone, as a feedback inhibitor on osteogenic differentiation. With anti-SOST as a potential therapy for osteoporosis in the near future, its basic biologic and phenotypic consequences in skeletal tumors should not be overlooked.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Neoplasias Ósseas/metabolismo , Neoplasias de Tecido Ósseo/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Fosfatase Alcalina/metabolismo , Biópsia , Neoplasias Ósseas/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Condroma/metabolismo , Condroma/patologia , Condrossarcoma/metabolismo , Condrossarcoma/patologia , Marcadores Genéticos , Humanos , Imuno-Histoquímica , Neoplasias de Tecido Ósseo/patologia , Osteoblastoma/metabolismo , Osteoblastoma/patologia , Osteogênese , Osteoma Osteoide/metabolismo , Osteoma Osteoide/patologia , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Estudos Retrospectivos , Via de Sinalização Wnt
11.
Hum Pathol ; 54: 92-9, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27063472

RESUMO

Pericytes are modified smooth muscle cells that closely enwrap small blood vessels, regulating and supporting the microvasculature through direct endothelial contact. Pericytes demonstrate a distinct immunohistochemical profile, including expression of smooth muscle actin, CD146, platelet-derived growth factor receptor ß, and regulator of G-protein signaling 5. Previously, pericyte-related antigens have been observed to be present among a group of soft tissue tumors with a perivascular growth pattern, including glomus tumor, myopericytoma, and angioleiomyoma. Similarly, malignant tumor cells have been shown to have a pericyte-like immunoprofile when present in a perivascular location, seen in malignant melanoma, glioblastoma, and adenocarcinoma. Here, we examine well-differentiated liposarcoma specimens, which showed some element of perivascular areas with the appearance of smooth muscle (n = 7 tumors). Immunohistochemical staining was performed for pericyte antigens, including smooth muscle actin, CD146, platelet-derived growth factor receptor ß, and regulator of G-protein signaling 5. Results showed consistent pericytic marker expression among liposarcoma tumor cells within a perivascular distribution. MDM2 immunohistochemistry and fluorescence in situ hybridization for MDM2 revealed that these perivascular cells were of tumor origin (7/7 tumors), whereas double immunohistochemical detection for CD31/CD146 ruled out an endothelial cell contribution. These findings further support the concept of pericytic mimicry, already established in diverse malignancies, and its presence in well-differentiated liposarcoma. The extent to which pericytic mimicry has prognostic significance in liposarcoma is as yet unknown.


Assuntos
Diferenciação Celular , Lipoma/patologia , Lipossarcoma/patologia , Mimetismo Molecular , Pericitos/patologia , Actinas/análise , Adulto , Idoso , Biomarcadores Tumorais/análise , Biópsia , Antígeno CD146/análise , Feminino , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Lipoma/química , Lipoma/genética , Lipossarcoma/química , Lipossarcoma/genética , Masculino , Pessoa de Meia-Idade , Pericitos/química , Fenótipo , Proteínas Proto-Oncogênicas c-mdm2/análise , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas RGS/análise , Receptor beta de Fator de Crescimento Derivado de Plaquetas/análise , Estudos Retrospectivos
12.
Tissue Eng Part B Rev ; 22(4): 284-97, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26857241

RESUMO

Bone morphogenetic protein-2 (BMP-2) is currently the only Food and Drug Administration (FDA)-approved osteoinductive growth factor used as a bone graft substitute. However, with increasing clinical use of BMP-2, a growing and well-documented side effect profile has emerged. This includes postoperative inflammation and associated adverse effects, ectopic bone formation, osteoclast-mediated bone resorption, and inappropriate adipogenesis. Several large-scale studies have confirmed the relative frequency of adverse events associated with the clinical use of BMP-2, including life-threatening cervical spine swelling. In fact, the FDA has issued a warning of the potential life-threatening complications of BMP-2. This review summarizes the known adverse effects of BMP-2, including controversial areas such as tumorigenesis. Next, select animal models that replicate BMP-2's adverse clinical effects are discussed. Finally, potential molecules to mitigate the adverse effects of BMP-2 are reviewed. In summary, BMP-2 is a potent osteoinductive cytokine that has indeed revolutionized the bone graft substitute market; however, it simultaneously has accrued a worrisome side effect profile. Better understanding of these adverse effects among both translational scientists and clinicians will help determine the most appropriate and safe use of BMP-2 in the clinical setting.


Assuntos
Proteína Morfogenética Óssea 2/efeitos adversos , Animais , Reabsorção Óssea , Humanos , Fusão Vertebral , Fator de Crescimento Transformador beta
13.
Am J Pathol ; 186(2): 419-34, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26772960

RESUMO

The differentiation factor NEL-like molecule-1 (NELL-1) has been reported as osteoinductive in multiple in vivo preclinical models. Bone morphogenetic protein (BMP)-2 is used clinically for skeletal repair, but in vivo administration can induce abnormal, adipose-filled, poor-quality bone. We demonstrate that NELL-1 combined with BMP2 significantly optimizes osteogenesis in a rodent femoral segmental defect model by minimizing the formation of BMP2-induced adipose-filled cystlike bone. In vitro studies using the mouse bone marrow stromal cell line M2-10B4 and human primary bone marrow stromal cells have confirmed that NELL-1 enhances BMP2-induced osteogenesis and inhibits BMP2-induced adipogenesis. Importantly, the ability of NELL-1 to direct BMP2-treated cells toward osteogenesis and away from adipogenesis requires intact canonical Wnt signaling. Overall, these studies establish the feasibility of combining NELL-1 with BMP2 to improve clinical bone regeneration and provide mechanistic insight into canonical Wnt pathway activity during NELL-1 and BMP2 osteogenesis. The novel abilities of NELL-1 to stimulate Wnt signaling and to repress adipogenesis may highlight new treatment approaches for bone loss in osteoporosis.


Assuntos
Adipogenia , Proteína Morfogenética Óssea 2/metabolismo , Regeneração Óssea/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Osteogênese/fisiologia , Animais , Proteínas de Ligação ao Cálcio , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Ratos Endogâmicos Lew , Transdução de Sinais/fisiologia
14.
Med Oncol ; 32(8): 210, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26123600

RESUMO

Perivascular epithelioid cell tumors (PEComas) are an uncommon family of soft tissue tumors with dual myoid-melanocytic differentiation. Although PEComa family tumors commonly demonstrate a perivascular growth pattern, pericyte antigen expression has not yet been examined among this unique tumor group. Previously, we demonstrated that a subset of perivascular soft tissue tumors exhibit a striking pericytic immunophenotype, with diffuse expression of αSMA, CD146, and PDGFRß. Here, we describe the presence of pericyte antigens across a diverse group of PEComa family tumors (n = 19 specimens). Results showed that pericyte antigens differed extensively by histological appearance. Typical angiomyolipoma (AML) specimens showed variable expression of pericyte antigens among both perivascular and myoid-appearing cells. In contrast, AML specimens with a predominant spindled morphology showed diffuse expression of pericyte markers, including αSMA, CD146, and PDGFRß. AML samples with predominant epithelioid morphology showed a marked reduction in or the absence of immunoreactivity for pericyte markers. Lymphangiomyoma samples showed more variable and partial pericyte marker expression. In summary, pericyte antigen expression is variable among PEComa family tumors and largely varies by tumor morphology. Pericytic marker expression in PEComa may represent a true pericytic cell of origin, or alternatively aberrant pericyte marker adoption. Markers of pericytic differentiation may be of future diagnostic utility for the evaluation of mesenchymal tumors, or identify actionable signaling pathways for future therapeutic intervention.


Assuntos
Angiomiolipoma/patologia , Antígenos de Neoplasias/análise , Pericitos/patologia , Neoplasias de Células Epitelioides Perivasculares/patologia , Actinas/análise , Actinas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Angiomiolipoma/imunologia , Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/análise , Antígeno CD146/análise , Antígeno CD146/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Pericitos/imunologia , Neoplasias de Células Epitelioides Perivasculares/imunologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/análise , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo
15.
Int J Surg Pathol ; 23(8): 638-48, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26085647

RESUMO

INTRODUCTION: Perivascular soft tissue tumors are relatively uncommon neoplasms of unclear line of differentiation, although most are presumed to originate from pericytes or modified perivascular cells. Among these, glomus tumor, myopericytoma, and angioleiomyoma share a spectrum of histologic findings and a perivascular growth pattern. In contrast, solitary fibrous tumor (previously termed hemangiopericytoma) was once hypothesized to have pericytic differentiation. METHODS: Here, we systematically examine pericyte immunohistochemical markers among glomus tumor (including malignant glomus tumor), myopericytoma, angioleiomyoma, and solitary fibrous tumor. Immunohistochemical staining and semiquantification was performed using well-defined pericyte antigens, including αSMA, CD146, and PDGFRß. RESULTS: Glomus tumor and myopericytoma demonstrate diffuse staining for all pericyte markers, including immunohistochemical reactivity for αSMA, CD146, and PDGFRß. Malignant glomus tumors all showed some degree of pericyte marker immunoreactivity, although it was significantly reduced. Angioleiomyoma shared a similar αSMA + CD146 + PDGFRß+ immunophenotype; however, this was predominantly seen in the areas of perivascular tumor growth. Solitary fibrous tumors showed patchy PDGFRß immunoreactivity only. DISCUSSION: In summary, pericyte marker expression is a ubiquitous finding in glomus tumor, myopericytoma, and angioleiomyoma. Malignant glomus tumor shows a comparative reduction in pericyte marker expression, which may represent partial loss of pericytic differentiation. Pericyte markers are essentially not seen in solitary fibrous tumor. The combination of αSMA, CD146, and PDGFRß immunohistochemical stainings may be of utility for the evaluation of pericytic differentiation in soft tissue tumors.


Assuntos
Antígenos de Neoplasias/biossíntese , Biomarcadores Tumorais/análise , Pericitos/patologia , Neoplasias de Tecidos Moles/patologia , Antígenos de Neoplasias/análise , Humanos , Imuno-Histoquímica , Pericitos/metabolismo , Estudos Retrospectivos , Neoplasias de Tecidos Moles/metabolismo
16.
J Orthop ; 12(Suppl 2): S223-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27047227

RESUMO

BACKGROUND/AIMS: NELL-1 is a novel osteochondral differentiation factor protein with increasing usage in tissue engineering. Previously, we reported the expression patterns of NELL-1 in bone-forming skeletal tumors. With increasing interest in the use of NELL-1 protein, we sought to examine the expression of NELL-1 in cartilage-forming tumors. METHODS: Immunohistochemical expression was examined in human pathologic specimens. RESULTS: Consistent NELL-1 overexpression across all cartilage-forming tumors was observed. Similar degrees of expression were observed in enchondroma, chondrosarcoma, and chondroblastic osteosarcoma. NELL-1 expression did not significantly vary by tumor grade. CONCLUSION: In summary, NELL-1 demonstrates reliable and consistent expression across cartilage-forming skeletal tumors.

17.
Med Oncol ; 32(1): 430, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25491143

RESUMO

Although rare, sarcomas represent a source of significant morbidity and mortality with nearly one reported death for every two new diagnoses. The detection and surveillance of circulating tumor cells (or CTCs) has been found to have significant clinical utility in epithelial malignancies, such as carcinoma of the colon, breast and prostate. Here, we summarize what is known regarding CTCs in sarcomas. Although still in its relative infancy, the detection of CTCs in sarcoma patients may help to diagnose and predict recurrence or metastasis as well as improve the overall management of sarcoma patients. CTCs are most often detected via reverse transcriptase polymerase chain reaction or antibody-based detection of cell surface proteins, including flow cytometry. Samples may be obtained from either peripheral blood or bone marrow. CTC detection in translocation sarcomas is perhaps most promising, as a recurrent abnormal gene fusion product can be detected in involved individuals but not in the normal patient. Studies in Ewing's sarcoma/peripheral neuroectodermal tumor, synovial sarcoma and alveolar soft part sarcoma have confirmed the feasibility of this approach. Other investigators have turned toward detection of more universal markers of sarcomas, such as the pan-mesenchymal marker Vimentin. In the case of osteosarcoma, more specific markers of osteogenic differentiation (Type I Collagen) have been utilized. In summary, although in its relative nascency, the use of CTC detection for the management of sarcoma patients shows initial promise.


Assuntos
Células Neoplásicas Circulantes/patologia , Sarcoma/sangue , Sarcoma/patologia , Humanos
18.
Stem Cells Transl Med ; 3(10): 1231-41, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25154782

RESUMO

Adipose tissue is an attractive source of mesenchymal stem cells (MSCs) because of its abundance and accessibility. We have previously defined a population of native MSCs termed perivascular stem cells (PSCs), purified from diverse human tissues, including adipose tissue. Human PSCs (hPSCs) are a bipartite cell population composed of pericytes (CD146+CD34-CD45-) and adventitial cells (CD146-CD34+CD45-), isolated by fluorescence-activated cell sorting and with properties identical to those of culture identified MSCs. Our previous studies showed that hPSCs exhibit improved bone formation compared with a sample-matched unpurified population (termed stromal vascular fraction); however, it is not known whether hPSCs would be efficacious in a spinal fusion model. To investigate, we evaluated the osteogenic potential of freshly sorted hPSCs without culture expansion and differentiation in a rat model of posterolateral lumbar spinal fusion. We compared increasing dosages of implanted hPSCs to assess for dose-dependent efficacy. All hPSC treatment groups induced successful spinal fusion, assessed by manual palpation and microcomputed tomography. Computerized biomechanical simulation (finite element analysis) further demonstrated bone fusion with hPSC treatment. Histological analyses showed robust endochondral ossification in hPSC-treated samples. Finally, we confirmed that implanted hPSCs indeed differentiated into osteoblasts and osteocytes; however, the majority of the new bone formation was of host origin. These results suggest that implanted hPSCs positively regulate bone formation via direct and paracrine mechanisms. In summary, hPSCs are a readily available MSC population that effectively forms bone without requirements for culture or predifferentiation. Thus, hPSC-based products show promise for future efforts in clinical bone regeneration and repair.


Assuntos
Transplante Ósseo/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Osteogênese/fisiologia , Fusão Vertebral/métodos , Animais , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Análise de Elementos Finitos , Xenoenxertos , Humanos , Imuno-Histoquímica , Ratos , Microtomografia por Raio-X
19.
Tissue Eng Part A ; 20(19-20): 2699-710, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24785569

RESUMO

INTRODUCTION: Skeletal aging is associated not only with alterations in osteoblast (OB) and osteoclast (OC) number and activity within the basic metabolic unit, but also with increased marrow adiposity. Peroxisome proliferator-activated receptor gamma (PPARγ) is commonly considered the master transcriptional regulator of adipogenesis, however, it has known roles in osteoblast and osteoclast function as well. Here, we designed a lentiviral delivery system for PPARγ shRNA, and examined its effects in vitro on bone marrow stromal cells (BMSC) and in a mouse intramedullary injection model. METHODS: PPARγ shRNA was delivered by a replication-deficient lentiviral vector, after in vitro testing to confirm purity, concentration, and efficacy for Pparg transcript reduction. Next, control green fluorescent protein lentivirus or PPARγ shRNA expressing lentivirus were delivered by intramedullary injection into the femoral bone marrow of male SCID mice. Analyses included daily monitoring of animal health, and postmortem analysis at 4 weeks. Postmortem analyses included high resolution microcomputed tomography (microCT) reconstructions and analysis, routine histology and histomorphometric analysis, quantitative real time polymerase chain reaction analysis of Pparg transcript levels, and immunohistochemical analysis for markers of adipocytes (PPARγ, fatty acid binding protein 4 [FABP4]), osteoblasts (alkaline phosphatase [ALP], osteocalcin [OCN]), and osteoclasts (tartrate-resistant acid phosphatase [TRAP], Cathepsin K). RESULTS: In vitro, PPARγ shRNA delivery significantly reduced Pparg expression in mouse BMSC, accompanied by a significant reduction in lipid droplet accumulation. In vivo, a near total reduction in mature marrow adipocytes was observed at 4 weeks postinjection. This was accompanied by significant reductions in adipocyte-specific markers. Parameters of trabecular bone were significantly increased by both microCT and histomorphometric analysis. By immunohistochemical staining and semi-quantification, a significant increase in OCN+osteoblasts and decrease in TRAP+multinucleated osteoclasts was observed with PPARγ shRNA treatment. DISCUSSION: These findings suggest that acute loss of PPARγ in the bone marrow compartment has a significant role beyond anti-adipose effects. Specifically, we found pro-osteoblastogenic, anti-osteoclastic effects after PPARγ shRNA treatment, resulting in improved trabecular bone architecture. Future studies will examine the isolated and direct effects of PPARγ shRNA on OB and OC cell types, and it may help determine whether PPARγ antagonists are potential therapeutic agents for osteoporotic bone loss.


Assuntos
Adipogenia , Fêmur/metabolismo , Técnicas de Transferência de Genes , Lentivirus , Osteogênese , PPAR gama/biossíntese , RNA Interferente Pequeno/biossíntese , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Fêmur/citologia , Regulação da Expressão Gênica/genética , Vetores Genéticos , Masculino , Camundongos , Camundongos SCID , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , PPAR gama/genética , RNA Interferente Pequeno/genética
20.
Int Orthop ; 38(9): 1819-24, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24566993

RESUMO

PURPOSE: Pericytes were once thought only to aid in angiogenesis and blood pressure control. Gradually, the known functions of pericytes and other perivascular stem cells (PSC) have broadly increased. The following review article will summarize the known functions and importance of pericytes across disciplines of pathology, stem cell biology, and tissue engineering. METHODS: A literature review was performed for studies examining the importance of pericytes in pathology, stem cell biology, and tissue engineering. RESULTS: The importance of pericytes most prominently includes the identification of the perivascular identity of mesenchymal stem cells (or MSC). Now, pericytes and other PSC are known to display surface markers and multilineage differentiation potential of MSC. Accordingly, interest in the purification and use of PSC for mesenchymal tissue formation and regeneration has increased. Significant demonstration of in vivo efficacy in bone and muscle regeneration has been made in laboratory animals. Contemporaneously with the uncovering of an MSC identity for pericytes, investigators in tumour biology have found biologically relevant roles for pericytes in tumor formation, lymphovascular invasion, and perivascular tumor spread. As well, the contribution of pericytes to perivascular tumors has been examined (and debated), including glomus tumour, myopericytoma and solitary fibrous tumour/hemangiopericytoma. In addition, an expanding recognition of pericyte mimicry and perivascular tumour invasion has occurred, encompassing common malignancies of the brain and skin. CONCLUSIONS: In summary, pericytes have a wide range of roles in health and disease. Pericytes are being increasingly studied for their role in tumour formation, growth and invasion. Likewise, the application of pericytes/PSC for mesenchymal tissue engineering is an expanding field of interest.


Assuntos
Neoplasias de Tecido Vascular/fisiopatologia , Patologia/tendências , Pericitos/fisiologia , Pesquisa com Células-Tronco , Engenharia Tecidual/tendências , Animais , Diferenciação Celular , Linhagem da Célula , Modelos Animais de Doenças , Humanos , Células-Tronco Mesenquimais , Invasividade Neoplásica/patologia , Invasividade Neoplásica/fisiopatologia , Neoplasias de Tecido Vascular/patologia , Pericitos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA