Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 44(7): 1601-1616, 2024 07.
Artigo em Inglês | MEDLINE | ID: mdl-38660803

RESUMO

BACKGROUND: RAB27A is a member of the RAS oncogene superfamily of GTPases and regulates cell secretory function. It, is expressed within blood vessels and perivascular adipose tissue. We hypothesized that loss of RAB27A would alter cardiovascular function. METHODS: Body weight of Rab27aash mice was measured from 2 to 18 months of age, along with glucose resorption at 6 and 12 months of age and glucose sensitivity at 18 months of age. Body weight and cellular and molecular features of perivascular adipose tissue and aortic tissue were examined in a novel C57BL/6J Rab27a null strain. Analyses included morphometric quantification and proteomic analyses. Wire myography measured vasoreactivity, and echocardiography measured cardiac function. Comparisons across ages and genotypes were evaluated via 2-way ANOVA with multiple comparison testing. Significance for myography was determined via 4-parameter nonlinear regression testing. RESULTS: Genome-wide association data linked rare human RAB27A variants with body mass index and glucose handling. Changes in glucose tolerance were observed in Rab27aash male mice at 18 months of age. In WT (wild-type) and Rab27a null male mice, body weight, adipocyte lipid area, and aortic area increased with age. In female mice, only body weight increased with age, independent of RAB27A presence. Protein signatures from male Rab27a null mice suggested greater associations with cardiovascular and metabolic phenotypes compared with female tissues. Wire myography results showed Rab27a null males exhibited increased vasoconstriction and reduced vasodilation at 8 weeks of age. Rab27a null females exhibited increased vasoconstriction and vasodilation at 20 weeks of age. Consistent with these vascular changes, male Rab27a null mice experienced age-related cardiomyopathy, with severe differences observed by 21 weeks of age. CONCLUSIONS: Global RAB27A loss impacted perivascular adipose tissue and thoracic aorta proteomic signatures, altered vasocontractile responses, and decreased left ventricular ejection fraction in mice.


Assuntos
Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas rab27 de Ligação ao GTP , Animais , Proteínas rab27 de Ligação ao GTP/genética , Proteínas rab27 de Ligação ao GTP/metabolismo , Masculino , Feminino , Camundongos , Fenótipo , Tecido Adiposo/metabolismo , Vasodilatação , Vasoconstrição , Fatores Etários , Proteômica , Fatores Sexuais , Aorta/metabolismo , Aorta/fisiopatologia , Humanos
2.
Cell Stress Chaperones ; 29(2): 312-325, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38490439

RESUMO

Type 1 diabetes (T1D) is characterized by lymphocyte infiltration into the pancreatic islets of Langerhans, leading to the destruction of insulin-producing beta cells and uncontrolled hyperglycemia. In the nonobese diabetic (NOD) murine model of T1D, the onset of this infiltration starts several weeks before glucose dysregulation and overt diabetes. Recruitment of immune cells to the islets is mediated by several chemotactic cytokines, including CXCL10, while other cytokines, including SDF-1α, can confer protective effects. Global gene expression studies of the pancreas from prediabetic NOD mice and single-cell sequence analysis of human islets from prediabetic, autoantibody-positive patients showed an increased expression of metallothionein (MT), a small molecular weight, cysteine-rich metal-binding stress response protein. We have shown that beta cells can release MT into the extracellular environment, which can subsequently enhance the chemotactic response of Th1 cells to CXCL10 and interfere with the chemotactic response of Th2 cells to SDF-1α. These effects can be blocked in vitro with a monoclonal anti-MT antibody, clone UC1MT. When administered to NOD mice before the onset of diabetes, UC1MT significantly reduces the development of T1D. Manipulation of extracellular MT may be an important approach to preserving beta cell function and preventing the development of T1D.


Assuntos
Diabetes Mellitus Tipo 1 , Estado Pré-Diabético , Humanos , Camundongos , Animais , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/prevenção & controle , Camundongos Endogâmicos NOD , Metalotioneína/genética , Metalotioneína/metabolismo , Quimiocina CXCL12
3.
J Vis Exp ; (145)2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30907879

RESUMO

Adipose tissue is a rich source of multi-potent mesenchymal stem cells (MSC) capable of differentiating into osteogenic, adipogenic, and chondrogenic lineages. Adipogenic differentiation of progenitor cells is a major mechanism driving adipose tissue expansion and dysfunction in response to obesity. Understanding changes to perivascular adipose tissue (PVAT) is thus clinically relevant in metabolic disease. However, previous studies have been predominately performed in the mouse and other animal models. This protocol uses human thoracic PVAT samples collected from patients undergoing coronary artery bypass graft surgery. Adipose tissue from the ascending aorta was collected and used for explantation of the stromal vascular fraction. We previously confirmed the presence of adipose progenitor cells in human PVAT with the capacity to differentiate into lipid-containing adipocytes. In this study, we further analyzed the differentiation potential of cells from the stromal vascular fraction, presumably containing multi-potent progenitor cells. We compared PVAT-derived cells to human bone marrow MSC for differentiation into adipogenic, osteogenic, and chondrogenic lineages. Following 14 days of differentiation, specific stains were utilized to detect lipid accumulation in adipocytes (Oil red O), calcific deposits in osteogenic cells (Alizarin Red), or glycosaminoglycans and collagen in chondrogenic cells (Masson's Trichrome). While bone marrow MSC efficiently differentiated into all three lineages, PVAT-derived cells had adipogenic and chondrogenic potential, but lacked robust osteogenic potential.


Assuntos
Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/citologia , Aorta/citologia , Diferenciação Celular , Adipócitos/citologia , Adipogenia , Animais , Forma Celular , Células Cultivadas , Condrogênese , Humanos , Células-Tronco Mesenquimais/citologia , Osteogênese , Células Estromais/citologia
4.
Artigo em Inglês | MEDLINE | ID: mdl-32133436

RESUMO

BACKGROUND: Adipose tissue is a vital tissue in mammals that functions to insulate our bodies, regulate our internal thermostat, protect our organs, store energy (and burn energy, in the case of beige and brown fat), and provide endocrine signals to other organs in the body. Tissue engineering of adipose and other soft tissues may prove essential for people who have lost this tissue from trauma or disease. MAIN TEXT: In this review, we discuss the applications of tissue-engineered adipose tissue specifically for disease modeling applications. We provide a basic background to adipose depots and describe three-dimensional (3D) in vitro adipose models for obesity, diabetes, and cancer research applications. CONCLUSIONS: The approaches to engineering 3D adipose models are diverse in terms of scaffold type (hydrogel-based, silk-based and scaffold-free), species of origin (H. sapiens and M. musculus) and cell types used, which allows researchers to choose a model that best fits their application, whether it is optimization of adipocyte differentiation or studying the interaction of adipocytes and other cell types like endothelial cells. In vitro 3D adipose tissue models support discoveries into the mechanisms of adipose-related diseases and thus support the development of novel anti-cancer or anti-obesity/diabetes therapies.

5.
Bone ; 118: 77-88, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29366838

RESUMO

Over the past twenty years, evidence has accumulated that biochemically and spatially defined networks of extracellular matrix, cellular components, and interactions dictate cellular differentiation, proliferation, and function in a variety of tissue and diseases. Modeling in vivo systems in vitro has been undeniably necessary, but when simplified 2D conditions rather than 3D in vitro models are used, the reliability and usefulness of the data derived from these models decreases. Thus, there is a pressing need to develop and validate reliable in vitro models to reproduce specific tissue-like structures and mimic functions and responses of cells in a more realistic manner for both drug screening/disease modeling and tissue regeneration applications. In adipose biology and cancer research, these models serve as physiologically relevant 3D platforms to bridge the divide between 2D cultures and in vivo models, bringing about more reliable and translationally useful data to accelerate benchtop to bedside research. Currently, no model has been developed for bone marrow adipose tissue (BMAT), a novel adipose depot that has previously been overlooked as "filler tissue" but has more recently been recognized as endocrine-signaling and systemically relevant. Herein we describe the development of the first 3D, BMAT model derived from either human or mouse bone marrow (BM) mesenchymal stromal cells (MSCs). We found that BMAT models can be stably cultured for at least 3 months in vitro, and that myeloma cells (5TGM1, OPM2 and MM1S cells) can be cultured on these for at least 2 weeks. Upon tumor cell co-culture, delipidation occurred in BMAT adipocytes, suggesting a bidirectional relationship between these two important cell types in the malignant BM niche. Overall, our studies suggest that 3D BMAT represents a "healthier," more realistic tissue model that may be useful for elucidating the effects of MAT on tumor cells, and tumor cells on MAT, to identify novel therapeutic targets. In addition, proteomic characterization as well as microarray data (expression of >22,000 genes) coupled with KEGG pathway analysis and gene set expression analysis (GSEA) supported our development of less-inflammatory 3D BMAT compared to 2D culture. In sum, we developed the first 3D, tissue-engineered bone marrow adipose tissue model, which is a versatile, novel model that can be used to study numerous diseases and biological processes involved with the bone marrow.


Assuntos
Tecido Adiposo/fisiologia , Medula Óssea/fisiologia , Modelos Biológicos , Animais , Linhagem Celular Tumoral , Feminino , Regulação da Expressão Gênica , Humanos , Lipídeos/isolamento & purificação , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/patologia , Proteômica , Seda/química , Engenharia Tecidual , Alicerces Teciduais/química
6.
Cell Rep ; 25(11): 3215-3228.e9, 2018 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-30540952

RESUMO

Molecular- and cellular-based therapies have the potential to reduce obesity-associated disease. In response to cold, beige adipocytes form in subcutaneous white adipose tissue and convert energy stored in metabolic substrates to heat, making them an attractive therapeutic target. We developed a robust method to generate a renewable source of human beige adipocytes from induced pluripotent stem cells (iPSCs). Developmentally, these cells are derived from FOXF1+ mesoderm and progress through an expandable mural-like mesenchymal stem cell (MSC) to form mature beige adipocytes that display a thermogenically active profile. This includes expression of uncoupling protein 1 (UCP1) concomitant with increased uncoupled respiration. With this method, dysfunctional adipogenic precursors can be reprogrammed and differentiated into beige adipocytes with increased thermogenic function and anti-diabetic secretion potential. This resource can be used to (1) elucidate mechanisms that underlie the control of beige adipogenesis and (2) generate material for cellular-based therapies that target metabolic syndrome in humans.


Assuntos
Adipócitos Bege/transplante , Síndrome Metabólica/terapia , Adipócitos Bege/citologia , Diferenciação Celular , Linhagem Celular , Fatores de Transcrição Forkhead/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Interleucina-4/farmacologia , Células-Tronco Mesenquimais/citologia , Mesoderma/citologia , Transdução de Sinais , Circulação Esplâncnica , Fator de Crescimento Transformador beta/metabolismo
7.
J Cell Biochem ; 119(4): 3267-3279, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29105817

RESUMO

Signals from growth factors or mechanical stimuli converge to promote vascular smooth muscle cell (VSMC) migration and proliferation, key events in the pathogenesis of intimal hyperplasia upon vascular injury. Spry1, a regulator of receptor tyrosine kinases (RTK), plays a role in maintaining the contractile phenotype of VSMC. The aim of the current study was to determine the role of Spry1 in VSMC proliferation in vitro and injury induced neointimal hyperplasia in vivo. VSMC proliferation and neointima formation were evaluated in cultured human aortic SMC (hAoSMC) and ligation-induced injury of mouse carotid arteries from Spry1 gene targeted mice, and their corresponding wild type littermates. Human Spry1 or non-targeting control lentiviral shRNAs were used to knock down Spry1 in hAoSMC. Time course cell cycle analysis showed a reduced fraction of S-phase cells at 12 and 24 h after growth medium stimulation in Spry1 shRNA transduced hAoSMC. Consistent with reduced S-phase entry, the induction of cyclinD1 and the levels of pRbS807/S811, pH3Ser10, and pCdc2 were also reduced, while the cell cycle inhibitor p27Kip1 was maintained in Spry1 knockdown hAoSMC. In vivo, loss of Spry1 attenuated carotid artery ligation-induced neointima formation in mice, and this effect was accompanied by a decrease in cell proliferation similar to the in vitro results. Our findings demonstrate that loss of Spry1 attenuates mitogen-induced VSMC proliferation, and thus injury-induced neointimal hyperplasia likely via insufficient activation of Akt signaling causing decreased cyclinD1 and increased p27Kip1 and a subsequent decrease in Rb and cdc2 phosphorylation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Lesões das Artérias Carótidas/complicações , Proteínas de Membrana/genética , Músculo Liso Vascular/citologia , Neointima/genética , Fosfoproteínas/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/metabolismo , Ciclo Celular , Proliferação de Células , Células Cultivadas , Ciclina D1/genética , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Músculo Liso Vascular/metabolismo , Neointima/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
8.
Cancer Cell Int ; 17: 53, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28507453

RESUMO

[This corrects the article DOI: 10.1186/s12935-016-0292-7.].

10.
Cancer Cell Int ; 16: 19, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26973433

RESUMO

BACKGROUND: Cancer stem cells contribute to tumor initiation, heterogeneity, and recurrence, and are critical targets in cancer therapy. Sprouty4 (Spry4) is a potent inhibitor of signal transduction pathways elicited by receptor tyrosine kinases, and has roles in regulating cell proliferation, migration and differentiation. Spry4 has been implicated as a tumor suppressor and in modulating embryonic stem cells. OBJECTIVES: The purpose of this research was to test the novel idea that Spry4 regulates cancer stem cell properties in breast cancer. METHODS: Loss-of function of Spry4 in human MDA-MB-231 cell was used to test our hypothesis. Spry4 knockdown or control cell lines were generated using lentiviral delivery of human Spry4 or non-targeting control shRNAs, and then selected with 2 µg/ml puromycin. Cell growth and migratory abilities were determined using growth curve and cell cycle flow cytometry analyses and scratch assays, respectively. Xenograft tumor model was used to determine the tumorigenic activity and metastasis in vivo. Cancer stem cell related markers were evaluated using immunoblotting assays and fluorescence-activated cell sorting. Cancer stem cell phenotype was evaluated using in vitro mammosphere formation and drug sensitivity tests, and in vivo limiting dilution tumor formation assay. RESULTS: Two out of three tested human Spry4 shRNAs significantly suppressed the expression of endogenous Spry4 in MDA-MB-231 cells. Suppressing Spry4 expression increased MDA-MB-231 cell proliferation and migration. Suppressing Spry4 increased ß3-integrin expression, and CD133(+)CD44(+) subpopulation. Suppressing Spry4 increased mammosphere formation, while decreasing the sensitivity of MDA-MB-231 cells to Paclitaxel treatment. Finally, suppressing Spry4 increased the potency of MDA-MB-231 cell tumor initiation, a feature attributed to cancer stem cells. CONCLUSIONS: Our findings provide novel evidence that endogenous Spry4 may have tumor suppressive activity in breast cancer by suppressing cancer stem cell properties in addition to negative effects on tumor cell proliferation and migration.

11.
Sci Rep ; 6: 23216, 2016 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-26976794

RESUMO

Sprouty (Spry) proteins have been implicated in cancer progression, but their role in triple-negative breast cancer (TNBC), a subtype of lethal and aggressive breast cancer, is unknown. Here, we reported that Spry1 is significantly expressed in TNBC specimen and MDA-MB-231 cells. To understand Spry1 regulation of signaling events controlling breast cancer phenotype, we used lentiviral delivery of human Spry1 shRNAs to suppress Spry1 expression in MDA-MB-231, an established TNBC cell line. Spry1 knockdown MDA-MB-231 cells displayed an epithelial phenotype with increased membrane E-cadherin expression. Knockdown of Spry1 impaired MDA-MB-231 cell migration, Matrigel invasion, and anchorage-dependent and -independent growth. Tumor xenografts originating from Spry1 knockdown MDA-MB-231 cells grew slower, had increased E-cadherin expression, and yielded fewer lung metastases compared to control. Furthermore, suppressing Spry1 in MDA-MB-231 cells impaired the induction of Snail and Slug expression by EGF, and this effect was associated with increased EGFR degradation and decreased EGFR/Grb2/Shp2/Gab1 signaling complex formation. The same phenotype was also observed in the TNBC cell line MDA-MB-157. Together, our results show that unlike in some tumors, where Spry may mediate tumor suppression, Spry1 plays a selective role in at least a subset of TNBC to promote the malignant phenotype via enhancing EGF-mediated mesenchymal phenotype.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Transição Epitelial-Mesenquimal , Receptores ErbB/metabolismo , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Colágeno , Combinação de Medicamentos , Feminino , Técnicas de Silenciamento de Genes , Humanos , Laminina , Proteínas de Membrana/genética , Camundongos Endogâmicos NOD , Camundongos SCID , Invasividade Neoplásica/patologia , Fosfoproteínas/genética , Proteoglicanas , Neoplasias de Mama Triplo Negativas/patologia
12.
Methods Mol Biol ; 1406: 255-69, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26820962

RESUMO

Models of tumor angiogenesis have played a critical role in understanding the mechanisms involved in the recruitment of vasculature to the tumor mass, and have also provided a platform for testing antiangiogenic potential of new therapeutics that combat the development of malignant growth. In this regard, the chorioallantoic membrane (CAM) of the developing chick embryo has proven to be an elegant model for investigation of angiogenic processes. Here, we describe methods for effectively utilizing the preestablished vascular network of the chick CAM to investigate and quantify tumor-associated angiogenesis in a breast tumor model.


Assuntos
Membrana Corioalantoide/irrigação sanguínea , Neovascularização Patológica , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Embrião de Galinha , Membrana Corioalantoide/diagnóstico por imagem , Membrana Corioalantoide/patologia , Humanos , Microtomografia por Raio-X
13.
Glia ; 63(12): 2198-207, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26148976

RESUMO

Infarcted regions of the brain after stroke are segregated from the intact brain by scar tissue comprising both fibrous and glial components. The extent and quality of scarring is influenced by inflammation. The matricellular glycoprotein osteopontin (OPN) is strongly induced in myeloid cells after stroke and may contribute to repair of ischemic brain lesions. To elucidate the role of OPN in scar formation, we induced photothrombotic brain infarction, characterized by circumscribed cortical infarctions with a well-defined border zone toward the intact brain parenchyma. The cellular source and functional role of OPN was addressed by studies in OPN null (OPN(-/-) ) mice, wild-type mice depleted of hematogenous monocytes/macrophages by clodronate-filled liposome treatment, and CCR2(-/-) bone marrow chimeric mice characterized by impaired hematogenous macrophage influx into the infarctions. OPN was mainly produced by hematogenous macrophages infiltrating into the inner border zone of the infarcts whereas astrocyte activation occurred in the outer border zone. In OPN(-/-) as well as macrophage-depleted mice, reactive astrocytes failed to properly extend processes from the periphery toward the center of the infarctions. This was associated with incomplete coverage of neovessels by astrocytic endfeet and persistent leakiness of the damaged blood brain barrier. In conclusion, OPN produced by hematogenous macrophages induces astrocyte process extension toward the infarct border zone, which may contribute to repair of the ischemic neurovascular unit.


Assuntos
Astrócitos/fisiologia , Barreira Hematoencefálica/fisiopatologia , Isquemia Encefálica/fisiopatologia , Macrófagos/metabolismo , Osteopontina/metabolismo , Acidente Vascular Cerebral/fisiopatologia , Animais , Aquaporina 4/metabolismo , Astrócitos/patologia , Barreira Hematoencefálica/patologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Isquemia Encefálica/patologia , Modelos Animais de Doenças , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteopontina/genética , Receptores CCR2/genética , Receptores CCR2/metabolismo , Acidente Vascular Cerebral/patologia
14.
PLoS One ; 10(4): e0122892, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25909848

RESUMO

Endoglin is a type III TGFß auxiliary receptor that is upregulated in endothelial cells during angiogenesis and, when mutated in humans, results in the vascular disease hereditary hemorrhagic telangiectasia (HHT). Though endoglin has been implicated in cell adhesion, the underlying molecular mechanisms are still poorly understood. Here we show endoglin expression in endothelial cells regulates subcellular localization of zyxin in focal adhesions in response to BMP9. RNA knockdown of endoglin resulted in mislocalization of zyxin and altered formation of focal adhesions. The mechanotransduction role of focal adhesions and their ability to transmit regulatory signals through binding of the extracellular matrix are altered by endoglin deficiency. BMP/TGFß transcription factors, SMADs, and zyxin have recently been implicated in a newly emerging signaling cascade, the Hippo pathway. The Hippo transcription coactivator, YAP1 (yes-associated protein 1), has been suggested to play a crucial role in mechanotransduction and cell-cell contact. Identification of BMP9-dependent nuclear localization of YAP1 in response to endoglin expression suggests a mechanism of crosstalk between the two pathways. Suppression of endoglin and YAP1 alters BMP9-dependent expression of YAP1 target genes CCN1 (cysteine-rich 61, CYR61) and CCN2 (connective tissue growth factor, CTGF) as well as the chemokine CCL2 (monocyte chemotactic protein 1, MCP-1). These results suggest a coordinate effect of endoglin deficiency on cell matrix remodeling and local inflammatory responses. Identification of a direct link between the Hippo pathway and endoglin may reveal novel mechanisms in the etiology of HHT.


Assuntos
Quimiocinas/metabolismo , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Fatores de Diferenciação de Crescimento/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Antígenos CD/metabolismo , Quimiocina CCL2/metabolismo , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Proteína Rica em Cisteína 61/metabolismo , Endoglina , Adesões Focais/metabolismo , Fator 2 de Diferenciação de Crescimento , Via de Sinalização Hippo , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Knockout , Modelos Biológicos , Fosfoproteínas/metabolismo , Receptores CCR2/metabolismo , Receptores de Superfície Celular/metabolismo , Proteínas Smad/metabolismo , Fatores de Transcrição , Proteínas de Sinalização YAP , Zixina/metabolismo
15.
Bone ; 75: 161-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25725265

RESUMO

Multiple myeloma is a B-cell malignancy characterized by the unrelenting proliferation of plasma cells. Multiple myeloma causes osteolytic lesions and fractures that do not heal due to decreased osteoblastic and increased osteoclastic activity. However, the exact relationship between osteoblasts and myeloma cells remains elusive. Understanding the interactions between these dynamic bone-forming cells and myeloma cells is crucial to understanding how osteolytic lesions form and persist and how tumors grow within the bone marrow. This review provides a comprehensive overview of basic and translational research focused on the role of osteoblasts in multiple myeloma progression and their relationship to osteolytic lesions. Importantly, current challenges for in vitro studies exploring direct osteoblastic effects on myeloma cells, and gaps in understanding the role of the osteoblast in myeloma progression are delineated. Finally, successes and challenges in myeloma treatment with osteoanabolic therapy (i.e., any treatment that induces increased osteoblastic number or activity) are enumerated. Our goal is to illuminate novel mechanisms by which osteoblasts may contribute to multiple myeloma disease progression and osteolysis to better direct research efforts. Ultimately, we hope this may provide a roadmap for new approaches to the pathogenesis and treatment of multiple myeloma with a particular focus on the osteoblast.


Assuntos
Mieloma Múltiplo/patologia , Osteoblastos/metabolismo , Progressão da Doença , Humanos
16.
Prostate ; 75(9): 907-16, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25728608

RESUMO

BACKGROUND: PTEN/AKT signaling plays a key role in prostate cancer development and maintenance of prostate cancer stem cells. How other oncogenes or tumor suppressors interact with this pathway remain to be elucidated. SLUG is an zinc finger transcription factor of the Snail superfamily, and it promotes cancer metastasis and determines the mammary stem cell state. METHODS: SLUG was overexpressed in cells by retroviral vector and knockdown of SLUG and PTEN was mediated by shRNAs-expressing lentiviruses. Expression level of SLUG and PTEN was examined by Western blot, RT-PCR, and qPCR analyses. PTEN promoter activity was measured by luciferase reporter assay. ChIP assay was used to measure the binding between SLUG and the PTEN promoter in vivo. RESULT: We showed that overexpression of SLUG decreased expression of PTEN tumor repressor in prostate cancer cell lines 22RV1 and DU145; conversely, knockdown of SLUG expression elevated PTEN expresson at both protein and RNA level in these cells. We demonstrated that SLUG overexpression inhibits PTEN promoter activity through the proximal promoter region in prostate cancer cells. By ChIP assay, we confirmed that SLUG directly binds to the PTEN promoter region covering the E-box sites. We also showed that Slug deficiency leads to an increased expression of PTEN in mouse embryo fibroblasts and prostate tissues. Importantly, we found that overexpression of SLUG increases drug resistance of DU145 prostate cancer cell line and knockdown of SLUG by shRNA sensitizes DU145 cell line to chemotherapeutic drugs. We further demonstrated that PTEN knockdown converts drug sensitivity of DU145 cells expressing SLUG shRNA to anticancer drugs. CONCLUSION: We provide compelling evidence showing that PTEN is a direct functional target of SLUG. Our findings offer new insight in the regulation of the PTEN/AKT pathway and provide a molecular basis for potential targeted therapies of prostate cancer Prostate 75:907-916, 2015. © 2015 Wiley Periodicals, Inc.


Assuntos
PTEN Fosfo-Hidrolase/biossíntese , Neoplasias da Próstata/metabolismo , Fatores de Transcrição/biossíntese , Animais , Western Blotting , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , PTEN Fosfo-Hidrolase/antagonistas & inibidores , PTEN Fosfo-Hidrolase/genética , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , RNA Neoplásico/química , RNA Neoplásico/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética
17.
Angiogenesis ; 18(1): 31-46, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25249331

RESUMO

A more complete understanding of the mechanisms that regulate the angiogenic switch, which contributes to the conversion of small dormant tumors to actively growing malignancies, is important for the development of more effective anti-angiogenic strategies for cancer therapy. While significant progress has been made in understanding the complex mechanisms by which integrin αvß3 expressed in endothelial cells governs angiogenesis, less is known concerning the ability of αvß3 expressed within the tumor cell compartment to modulate the angiogenic output of a tumor. Here we provide evidence that αvß3 expressed in melanoma cells may contribute to the suppression of IGFBP-4, an important negative regulator of IGF-1 signaling. Given the multiple context-dependent roles for αvß3 in angiogenesis and tumor progression, our novel findings provide additional molecular insight into how αvß3 may govern the angiogenic switch by a mechanism associated with a p38 MAPK and matrix metalloproteinases-dependent regulation of the endogenous angiogenesis inhibitor IGFBP-4.


Assuntos
Proteína 4 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Integrina alfaVbeta3/antagonistas & inibidores , Melanoma/fisiopatologia , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Primers do DNA/genética , Imunofluorescência , Humanos , Imuno-Histoquímica , Melanoma/complicações , Neovascularização Patológica/etiologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Microtomografia por Raio-X
18.
PLoS One ; 8(3): e58746, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23554919

RESUMO

BACKGROUND: Changes in the vascular smooth muscle cell (VSMC) contractile phenotype occur in pathological states such as restenosis and atherosclerosis. Multiple cytokines, signaling through receptor tyrosine kinases (RTK) and PI3K/Akt and MAPK/ERK pathways, regulate these phenotypic transitions. The Spry proteins are feedback modulators of RTK signaling, but their specific roles in VSMC have not been established. METHODOLOGY/PRINCIPAL FINDINGS: Here, we report for the first time that Spry1, but not Spry4, is required for maintaining the differentiated state of human VSMC in vitro. While Spry1 is a known MAPK/ERK inhibitor in many cell types, we found that Spry1 has little effect on MAPK/ERK signaling but increases and maintains Akt activation in VSMC. Sustained Akt signaling is required for VSMC marker expression in vitro, while ERK signaling negatively modulates Akt activation and VSMC marker gene expression. Spry4, which antagonizes both MAPK/ERK and Akt signaling, suppresses VSMC differentiation marker gene expression. We show using siRNA knockdown and ChIP assays that FoxO3a, a downstream target of PI3K/Akt signaling, represses myocardin promoter activity, and that Spry1 increases, while Spry4 decreases myocardin mRNA levels. CONCLUSIONS: Together, these data indicate that Spry1 and Spry4 have opposing roles in VSMC phenotypic modulation, and Spry1 maintains the VSMC differentiation phenotype in vitro in part through an Akt/FoxO/myocardin pathway.


Assuntos
Aorta/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transativadores/metabolismo , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Expressão Gênica , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Miócitos de Músculo Liso/citologia , Proteínas do Tecido Nervoso/genética , Fenótipo , Fosfoproteínas/genética , Transdução de Sinais , Transcrição Gênica
19.
Transgenic Res ; 22(2): 403-10, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22851140

RESUMO

Hematopoietic stem cells (HSC) are found in several independent sites embryonically. Loss-of-function studies indicated that Notch1, but not Notch2 signaling was required for HSC emergence from the aortic-gonado-mesonephros (AGM) region. We previously showed that constitutive Notch1 activation impaired primitive erythroid differentiation, but its effects on HSC emergence from the AGM region were not studied. To further define specific roles of Notch receptors, we characterized HSC in mouse embryos expressing either Notch1 intracellular domain (ICD) or Notch4ICD in VE-cadherin or SM22α expressing populations. Although embryonic Notch1 activation in VE-cadherin populations led to lethality after E13.5, earlier defects in the fetal liver were observed. Embryos were analyzed at E12.5 to assess hematopoiesis and the phenotype of developing cells in the AGM region. We found that activation of Notch1 in the endothelial compartment in VE-cadherin expressing cells resulted in the absence of intra-aortic clusters and defects in fetal liver hematopoiesis. In contrast, although Notch4 expression is regulated during fetal hematopoiesis, activation of Notch4 in VE-cadherin expressing populations did not affect HSC phenotype, although later vascular remodeling was impaired. Likewise, activation of Notch1 in SM22α positive populations had no significant effect on hematopoiesis. Our results indicate a cell type-dependent activity and distinct features of Notch1 versus Notch4 signaling and their impact on HSC generation.


Assuntos
Antígenos CD/genética , Caderinas/genética , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Receptor Notch1/genética , Animais , Aorta/citologia , Aorta/embriologia , Aorta/metabolismo , Diferenciação Celular/genética , Células Endoteliais/citologia , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/metabolismo , Fígado/citologia , Fígado/embriologia , Fígado/crescimento & desenvolvimento , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas/genética , Receptor Notch4 , Receptores Notch/genética , Receptores Notch/metabolismo , Ativação Transcricional
20.
Blood ; 120(20): 4263-73, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23018639

RESUMO

BMP9 signaling has been implicated in hereditary hemorrhagic telangiectasia (HHT) and vascular remodeling, acting via the HHT target genes, endoglin and ALK1. This study sought to identify endothelial BMP9-regulated proteins that could affect the HHT phenotype. Gene ontology analysis of cDNA microarray data obtained after BMP9 treatment of primary human endothelial cells indicated regulation of chemokine, adhesion, and inflammation pathways. These responses included the up-regulation of the chemokine CXCL12/SDF1 and down-regulation of its receptor CXCR4. Quantitative mass spectrometry identified additional secreted proteins, including the chemokine CXCL10/IP10. RNA knockdown of endoglin and ALK1 impaired SDF1/CXCR4 regulation by BMP9. Because of the association of SDF1 with ischemia, we analyzed its expression under hypoxia in response to BMP9 in vitro, and during the response to hindlimb ischemia, in endoglin-deficient mice. BMP9 and hypoxia were additive inducers of SDF1 expression. Moreover, the data suggest that endoglin deficiency impaired SDF1 expression in endothelial cells in vivo. Our data implicate BMP9 in regulation of the SDF1/CXCR4 chemokine axis in endothelial cells and point to a role for BMP9 signaling via endoglin in a switch from an SDF1-responsive autocrine phenotype to an SDF1 nonresponsive paracrine state that represses endothelial cell migration and may promote vessel maturation.


Assuntos
Células Endoteliais/citologia , Fatores de Diferenciação de Crescimento/fisiologia , Neovascularização Fisiológica/fisiologia , Receptores de Ativinas Tipo I/fisiologia , Receptores de Activinas Tipo II/fisiologia , Animais , Antígenos CD/fisiologia , Aorta/citologia , Comunicação Autócrina , Hipóxia Celular , Movimento Celular , Quimiocina CXCL12/biossíntese , Quimiocina CXCL12/metabolismo , Meios de Cultivo Condicionados , Endoglina , Células Endoteliais/efeitos dos fármacos , Fator 2 de Diferenciação de Crescimento/farmacologia , Fator 2 de Diferenciação de Crescimento/fisiologia , Membro Posterior/irrigação sanguínea , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Isquemia/fisiopatologia , Camundongos , Comunicação Parácrina , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Superfície Celular/fisiologia , Fator de Crescimento Transformador beta1/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA