Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 5(1): 1145, 2022 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-36307522

RESUMO

Diabetes affects select organs such as the eyes, kidney, heart, and brain. Our recent studies show that diabetes also enhances adipogenesis in the bone marrow and reduces the number of marrow-resident vascular regenerative stem cells. In the current study, we have performed a detailed spatio-temporal examination to identify the early changes that are induced by diabetes in the bone marrow. Here we show that short-term diabetes causes structural and molecular changes in the marrow, including enhanced adipogenesis in tibiae of mice, prior to stem cell depletion. This enhanced adipogenesis was associated with suppressed transforming growth factor-beta (TGFB) signaling. Using human bone marrow-derived mesenchymal progenitor cells, we show that TGFB pathway suppresses adipogenic differentiation through TGFB-activated kinase 1 (TAK1). These findings may inform the development of novel therapeutic targets for patients with diabetes to restore regenerative stem cell function.


Assuntos
Diabetes Mellitus , Células-Tronco Mesenquimais , Humanos , Camundongos , Animais , Medula Óssea/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Células-Tronco Mesenquimais/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Fatores de Crescimento Transformadores/metabolismo
2.
Int Immunopharmacol ; 112: 109218, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36116148

RESUMO

BACKGROUND: Kidney damage is a frequent event in the course of hypertension. Recent researches highlighted a critical role of non-hemodynamic activities of angiotensin II (Ang II) in hypertension-associated kidney fibrosis and inflammation. These activities are mediated through toll-like receptors (TLRs) but the mechanisms by which Ang II links TLRs to downstream inflammatory and fibrogenic responses is not fully known. In this study, we investigated the role of TLR adapter protein called myeloid differentiation primary-response protein-88 (MyD88) as the potential link. METHODS: C57BL/6 mice were administered Ang II by micro-osmotic pump infusion for 4 weeks to develop nephropathy. Mice were treated with small-molecule MyD88 inhibitor LM8. In vitro, MyD88 was blocked using siRNA or LM8 in Ang II-challenged renal tubular epithelial cells. RESULTS: We show that MyD88 is mainly located in tubular epithelial cells and Ang II increases the interaction between TLR4 and MyD88. This interaction activates MAPKs and nuclear factor-κB (NF-κB), leading to increased production of inflammatory and fibrogenic factors. Inhibition of MyD88 by siRNA or selective inhibitor LM8 supresses MyD88-TLR4 interaction, NF-κB activation, and elaboration of inflammatory cytokines and fibrosis-associated factors. These protective actions resulted in decreased renal pathological changes and preserved renal function in LM8-treated hypertensive mice, without affecting hypertension. CONCLUSION: These results demonstrate that Ang II induces inflammation and fibrosis in renal tubular epithelial cells through MyD88 and present MyD88 as a potential point of intervention for hypertension-associated kidney disease.


Assuntos
Hipertensão Renal , Hipertensão , Camundongos , Animais , Angiotensina II/metabolismo , NF-kappa B/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Receptor 4 Toll-Like/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Camundongos Endogâmicos C57BL , Transdução de Sinais , Hipertensão Renal/induzido quimicamente , Hipertensão Renal/tratamento farmacológico , Hipertensão Renal/metabolismo , Rim/patologia , Fibrose , Citocinas/metabolismo , Inflamação/metabolismo , Hipertensão/induzido quimicamente , Hipertensão/tratamento farmacológico , Hipertensão/metabolismo
3.
Adv Sci (Weinh) ; 9(31): e2202590, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36180407

RESUMO

Diabetes manifests as chronic inflammation and leads to the development diabetic cardiomyopathy (DCM). Targeting key proteins in inflammatory signaling may provide new therapy for DCM. In this study, the authors explore the pharmacological effects and mechanisms of Schisandrin B (Sch B), a natural compound with anti-inflammatory activity against DCM. It is shown that Sch B prevents high-level glucose (HG)-induced hypertrophic and fibrotic responses in cultured cardiomyocytes. RNA sequencing and inflammatory qPCR microarray show that Sch B mainly affects myeloid differentiation primary response 88 (MyD88)-dependent inflammatory gene expression in HG-challenged cardiomyocytes. Further studies indicate that Sch B directly binds to and inhibits MyD88 activation, but does not alter MyD88-independent Toll-like receptor signaling in vivo and in vitro. Inhibiting or silencing MyD88 is associated with reduced levels of HG-induced inflammatory cytokines and myocardial injuries in vitro. Treatment of type 1 and type 2 diabetic mice with Sch B protects heart function, reduces myocardial injuries, and decreases secretion of inflammatory cytokines. Cardiomyocyte-specific MyD88 knockout also protects mice against cardiac inflammation and injury in type 1 diabetic mice. In conclusion, these studies show that cardiomyocyte MyD88 plays an apathogenetic role in DCM and Sch B specifically targets MyD88 to reduce inflammatory DCM.


Assuntos
Diabetes Mellitus Experimental , Cardiomiopatias Diabéticas , Animais , Camundongos , Citocinas/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/complicações , Cardiomiopatias Diabéticas/tratamento farmacológico , Cardiomiopatias Diabéticas/complicações , Cardiomiopatias Diabéticas/prevenção & controle , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/uso terapêutico
4.
Hypertension ; 79(9): 2028-2041, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35862110

RESUMO

BACKGROUND: Elevated Ang II (angiotensin II) level leads to a range of conditions, including hypertensive kidney disease. Recent evidences indicate that FGFR1 (fibroblast growth factor receptor 1) signaling may be involved in kidney injuries. In this study, we determined whether Ang II alters FGFR1 signaling to mediate renal dysfunction. METHODS: Human archival kidney samples from patients with or without hypertension were examined. Multiple genetic and pharmacological approaches were used to investigate FGFR1-mediated signaling in tubular epithelial NRK-52E cells in response to Ang II stimulation. C57BL/6 mice were infused with Ang II for 28 days to develop hypertensive kidney disease. Mice were treated with either adeno-associated virus expressing FGFR1 shRNA or FGFR1 inhibitor AZD4547. RESULTS: Kidney specimens from subjects with hypertension and mice challenged with Ang II have increased FGFR1 activity in renal epithelial cells. Renal epithelial cells in culture initiate extracellular matrix programming in response to Ang II, through the activation of FGFR1, which is independent of both AT1R (angiotensin II receptor type 1) and AT2R (angiotensin II receptor type 2). The RNA sequencing analysis indicated that disrupting FGFR1 suppresses Ang II-induced fibrogenic responses in epithelial cells. Mechanistically, Ang II-activated FGFR1 leads to STAT3 (signal transducer and activator of transcription 3) activation, which is responsible for fibrogenic factor expression in kidneys. In the mouse model of hypertensive kidney disease, genetic knockdown of FGFR1 or pharmacological inhibition of its activity protected kidneys from dysfunction and fibrosis upon Ang II challenge. CONCLUSIONS: Our studies uncover a novel mechanism causing renal fibrosis in hypertension and indicate FGFR1 as a potential target to preserve renal function and integrity.


Assuntos
Hipertensão Renal , Hipertensão , Angiotensina II/metabolismo , Angiotensina II/farmacologia , Animais , Células Epiteliais/metabolismo , Fibrose , Humanos , Hipertensão/metabolismo , Hipertensão Renal/metabolismo , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nefrite , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptores de Angiotensina/metabolismo
5.
Nat Commun ; 11(1): 2148, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32358497

RESUMO

Hyperglycemia activates toll-like receptor 4 (TLR4) to induce inflammation in diabetic cardiomyopathy (DCM). However, the mechanisms of TLR4 activation remain unclear. Here we examine the role of myeloid differentiation 2 (MD2), a co-receptor of TLR4, in high glucose (HG)- and diabetes-induced inflammatory cardiomyopathy. We show increased MD2 in heart tissues of diabetic mice and serum of human diabetic subjects. MD2 deficiency in mice inhibits TLR4 pathway activation, which correlates with reduced myocardial remodeling and improved cardiac function. Mechanistically, we show that HG induces extracellular advanced glycation end products (AGEs), which bind directly to MD2, leading to formation of AGEs-MD2-TLR4 complex and initiation of pro-inflammatory pathways. We further detect elevated AGE-MD2 complexes in heart tissues and serum of diabetic mice and human subjects with DCM. In summary, we uncover a new mechanism of HG-induced inflammatory responses and myocardial injury, in which AGE products directly bind MD2 to drive inflammatory DCM.


Assuntos
Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/metabolismo , Cardiomiopatias Diabéticas/imunologia , Cardiomiopatias Diabéticas/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Animais , Western Blotting , Calorimetria , Linhagem Celular , Humanos , Imunoprecipitação , Antígeno 96 de Linfócito/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/metabolismo
6.
Circ Res ; 126(8): 1007-1023, 2020 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-32098592

RESUMO

RATIONALE: Excessive Ang II (angiotensin II) levels lead to a profibrotic and hypertrophic milieu that produces deleterious remodeling and dysfunction in hypertension-associated heart failure. Agents that disrupt Ang II-induced cardiac dysfunction may have clinical utility in the treatment of hypertension-associated heart failure. OBJECTIVE: We have examined the potential effect of celastrol-a bioactive compound derived from the Celastraceae family-on Ang II-induced cardiac dysfunction. METHODS AND RESULTS: In rat primary cardiomyocytes and H9C2 (rat cardiomyocyte-like H9C2) cells, celastrol attenuates Ang II-induced cellular hypertrophy and fibrotic responses. Proteome microarrays, surface plasmon resonance, competitive binding assays, and molecular simulation were used to identify the molecular target of celastrol. Our data showed that celastrol directly binds to and inhibits STAT (signal transducer and activator of transcription)-3 phosphorylation and nuclear translocation. Functional tests demonstrated that the protection of celastrol is afforded through targeting STAT3. Overexpression of STAT3 dampens the effect of celastrol by partially rescuing STAT3 activity. Finally, we investigated the in vivo effect of celastrol treatment in mice challenged with Ang II and in the transverse aortic constriction model. We show that celastrol administration protected heart function in Ang II-challenged and transverse aortic constriction-challenged mice by inhibiting cardiac fibrosis and hypertrophy. CONCLUSIONS: Our studies show that celastrol inhibits Ang II-induced cardiac dysfunction by inhibiting STAT3 activity.


Assuntos
Angiotensina II/toxicidade , Sistemas de Liberação de Medicamentos/métodos , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Triterpenos/administração & dosagem , Remodelação Ventricular/efeitos dos fármacos , Animais , Linhagem Celular , Células HEK293 , Humanos , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Triterpenos Pentacíclicos , Estrutura Terciária de Proteína , Distribuição Aleatória , Ratos , Fator de Transcrição STAT3/química , Tripterygium , Triterpenos/química , Remodelação Ventricular/fisiologia
7.
Biochim Biophys Acta Mol Basis Dis ; 1866(5): 165683, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31953218

RESUMO

Arachidonic acid (AA) plays a fundamental role in the function of all cells. Metabolites of AA contribute to inflammation as well as for resolving inflammation. Although AA-derived metabolites exhibit well-substantiated bioactivity, it is not known whether AA regulates inflammatory responses independent of its metabolites. With the recent discovery that saturated fatty acids activate toll-like receptor-4 (TLR4), we tested the hypothesis that AA directly regulates inflammatory responses through modulating the activity of TLR4. In cultured cardiomyocytes and macrophages, we found that AA prevents saturated fatty acid-induced TLR4 complex formation with accessory proteins and the induction of proinflammatory cytokines. We discovered that AA directly binds to TLR4 co-receptor, myeloid differentiation factor 2 (MD2) and prevents saturated fatty acids from activating TLR4 pro-inflammatory signaling pathway. Similarly, AA reduced lipopolysaccharide (LPS)-induced inflammation in macrophages and septic death in mice through binding to MD2. In high-fat diet mouse model of obesity and LPS-induced model of acute lung injury, both mediating inflammatory responses through TLR4, treatment with AA prevented MD2/TLR4 dimerization, induction of inflammatory factors, and tissue injuries. In summary, we have discovered that AA interacts with MD2 and disrupts TLR4 activation by LPS and saturated fatty acids. These findings provide experimental evidence for a direct mechanism of AA-induced regulation of inflammation.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Ácido Araquidônico/farmacologia , Miocardite/tratamento farmacológico , Obesidade/complicações , Sepse/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Lesão Pulmonar Aguda/imunologia , Animais , Ácido Araquidônico/uso terapêutico , Linhagem Celular , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Ácidos Graxos/imunologia , Ácidos Graxos/metabolismo , Humanos , Lipopolissacarídeos/imunologia , Pulmão/imunologia , Pulmão/patologia , Antígeno 96 de Linfócito/antagonistas & inibidores , Antígeno 96 de Linfócito/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Miocardite/imunologia , Miocardite/patologia , Miocárdio/imunologia , Miocárdio/patologia , Miócitos Cardíacos , Obesidade/imunologia , Obesidade/metabolismo , Ácido Palmítico/toxicidade , Cultura Primária de Células , Ratos , Sepse/imunologia , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/metabolismo
8.
Oral Surg Oral Med Oral Pathol Oral Radiol ; 129(3): 246-259.e1, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31902664

RESUMO

OBJECTIVE: The aim of this study was to determine the utility of surrogate markers of human papillomavirus (HPV) infection in the diagnosis of HPV-associated oral epithelial dysplasia (OED). STUDY DESIGN: Twelve cases of oral dysplasia with histologic features of HPV infection were stained with surrogate markers for HPV (p16, Ki-67, and ProExC) on immunohistochemistry. A second group of 12 cases of oral dysplasia without histologic features of HPV infection was used for comparison. Reverse transcriptase polymerase chain reaction (RT-PCR) was used to confirm the presence of high-risk HPV (HR HPV) in p16-positive cases. RESULTS: All of the surrogate markers showed a statistically significant association with HPV-positive OED (P < .001). The agreement between p16 and HPV positivity was the strongest (κ = 1.00), whereas Ki-67 showed very good association with HPV (κ = 0.83), and ProExC showed good association (κ = 0.75). In each case, the agreement was statistically significant (P < .001). Overall, each of the 3 markers showed good sensitivity; however, ProExC showed the lowest specificity. CONCLUSIONS: The clinicopathologic features of 12 cases of HPV OED are reported. Diffuse p16 positivity is an accurate and reliable method for predicting HR HPV infection in both high and low grade cases of epithelial dysplasia with histopathologic features of HPV OED. The use of Ki-67 and ProExC did not demonstrate any additional diagnostic benefit in the diagnosis HPV OED.


Assuntos
Carcinoma in Situ , Papillomaviridae , Infecções por Papillomavirus , Biomarcadores Tumorais , Inibidor p16 de Quinase Dependente de Ciclina , Humanos , Imuno-Histoquímica , Antígeno Ki-67
9.
Head Neck Pathol ; 13(3): 286-297, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30120721

RESUMO

Pleomorphic adenoma (PA) is the most common benign salivary gland tumor. Kallikrein-related peptidases have been identified as biomarkers in many human tumors and may influence tumor behavior. We investigated KLK1-15 messenger ribonucleic acid and proteins in PA specimens to determine a KLK expression profile for this tumor. Fresh frozen PA tissue specimens (n = 26) and matched controls were subjected to quantitative real-time reverse transcription polymerase chain reaction to detect KLK1-15 mRNA. Expression of KLK1, KLK12, KLK13, and KLK8 proteins were then evaluated via immunostaining techniques. Statistical analyses were performed with the level of significance set at P < .05. We observed downregulation of KLK1, KLK12, and KLK13 mRNA expression, and immunostaining studies revealed downregulation of the corresponding proteins. Histologic evidence of capsular perforation was associated with increased KLK1 protein expression. Tumor size was not associated with capsular invasion and/or perforation. This study is the first to detail a KLK expression profile for PA at both the transcriptional level and the protein level. Future work is required to develop clinical applications of these findings.


Assuntos
Adenoma Pleomorfo/patologia , Biomarcadores Tumorais/análise , Calicreínas/análise , Neoplasias das Glândulas Salivares/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
10.
Artigo em Inglês | MEDLINE | ID: mdl-31920992

RESUMO

Chronic inflammation and oxidative stress lead to a multitude of adverse cellular responses in target organs of chronic diabetic complications. Curcumin, a highly investigated phytochemical, has been shown to exhibit both anti-inflammatory and antioxidant activities. However, the clinical application of curcumin has been greatly limited due to a poor pharmacokinetic profile. To overcome these limitations, we have generated analogs of curcumin to enhance bioavailability and offer a preferable pharmacokinetic profile. Here, we explored the effects of two mono-carbonyl curcumin analogs, L2H21 and L50H46, in alleviating indices of inflammation and oxidative stress in cell culture and mouse model of diabetic complications. Our results show that L2H21 and L50H46 normalize inflammatory mediators (IL-6 and TNF-α), extracellular matrix proteins (FN and COL4α1), vasoactive factors (VEGF and ET-1) and a key transcriptional coactivator (p300) in cultured human retinal microvascular endothelial cells (HRECs) and dermal-derived microvascular endothelial cells (HMVECs) challenged with high levels of glucose. These curcumin analogs also reduced glucose-induced oxidative DNA damage as evidenced by 8-OHdG labeling. We further show that treatment of streptozotocin-induced diabetic mice with curcumin analogs prevents cardiac and renal dysfunction. The preservation of target tissue function was associated with normalization of pro-inflammatory cytokines and matrix proteins. Collectively, our results show that L2H21 and L50H46 offer the anti-inflammatory and antioxidant activities as has been reported for curcumin, and may provide a clinically applicable therapeutic option for the treatment of diabetic complications.

11.
Oncotarget ; 8(56): 96263-96275, 2017 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-29221204

RESUMO

11ß-HSD1 has been recognized as a potential therapeutic target for type 2 diabetes. Recent studies have shown that hyperglycemia leads to activation of 11ß-HSD1, increasing the intracellular glucocorticoid levels. Excess glucocorticoids may lead to the clinical manifestations of cardiac injury. Therefore, the aim of this study is to investigate whether 11ß-HSD1 activation contributes to the development of diabetic cardiomyopathy. To investigate the role of 11ß-HSD1, we administered a selective 11ß-HSD1 inhibitor in type 1 and type 2 murine models of diabetes and in cultured cardiomyocytes. Our results show that diabetes increases cortisone levels in heart tissues. 11ß-HSD1 inhibitor decreased cortisone levels and ameliorated all structural and functional features of diabetic cardiomyopathy including fibrosis and hypertrophy. We also show that high levels of glucose caused cardiomyocyte hypertrophy and increased matrix protein deposition in culture. Importantly, inhibition of 11ß-HSD1 attenuated these changes. Moreover, we show that 11ß-HSD1 activation mediates these changes through modulating EGFR phosphorylation and activity. Our findings demonstrate that 11ß-HSD1 contributes to the development of diabetic cardiomyopathy through activation of glucocorticoid and EGFR signaling pathway. These results suggest that inhibition of 11ß-HSD1 might be a therapeutic strategy for diabetic cardiomyopathy, which is independent of its effects on glucose homeostasis.

12.
J Mol Cell Cardiol ; 79: 1-12, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25444713

RESUMO

Obesity and increased free fatty acid (FFA) level are tightly linked, leading to the development of cardiovascular disorders. Curcumin is a natural product from Curcuma longa with multiple bioactivities and is known to have cardioprotective effects in several cellular and animal models. The current study was designed to evaluate the cardioprotective effects of curcumin and demonstrate the underlying mechanism in FFA-induced cardiac injury. Using cell culture studies and high fat in vivo model, we explored the mechanistic basis of anti-inflammatory and antioxidant activities of curcumin. We observed that palmitate (PA) treatment in cardiac derived H9C2 cells induced a marked increase in reactive oxygen species, inflammation, apoptosis and hypertrophy. All of these changes were effectively suppressed by curcumin treatment. In addition, oral administration of curcumin at 50mg/kg completely suppressed high fat diet-induced oxidative stress, inflammation, apoptosis, fibrosis, hypertrophy and tissue remodeling in mice. The beneficial actions of curcumin are closely associated with its ability to increase Nrf2 expression and inhibit NF-κB activation. Thus, both in vitro and in vivo studies showed a promising role of curcumin as a cardioprotective agent against palmitate and high fat diet mediated cardiac dysfunction. We indicated the regulatory roles of Nrf2 and NF-κB in obesity-induced heart injury, and suggested that they may be important therapeutic targets in the treatment of obesity-related disorders.


Assuntos
Cardiotônicos/uso terapêutico , Curcumina/farmacologia , Ácidos Graxos não Esterificados/efeitos adversos , Miocárdio/metabolismo , Miocárdio/patologia , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Cardiomegalia/complicações , Cardiomegalia/tratamento farmacológico , Cardiomegalia/patologia , Cardiotônicos/farmacologia , Linhagem Celular , Curcumina/administração & dosagem , Curcumina/uso terapêutico , Dieta Hiperlipídica , Fibrose , Masculino , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , Palmitatos/efeitos adversos , Ratos , Espécies Reativas de Oxigênio/metabolismo
13.
Stem Cells ; 32(6): 1649-60, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24496952

RESUMO

Human bone marrow mesenchymal progenitor cells (MPCs) are multipotent cells that play an essential role in endogenous repair and the maintenance of the stem cell niche. We have recently shown that high levels of glucose, conditions mimicking diabetes, cause impairment of MPCs, resulting in enhanced adipogenesis and suppression of osteogenesis. This implies that diabetes may lead to reduced endogenous repair mechanisms through altering the differentiation potential of MPCs and, consequently, disrupting the stem cell niche. Phenotypic alterations in the bone marrow of long-term diabetic patients closely resemble this observation. Here, we show that high levels of glucose selectively enhance autogenous Wnt11 expression in MPCs to stimulate adipogenesis through the Wnt/protein kinase C noncanonical pathway. This novel mechanism may account for increased bone marrow adipogenesis, severe bone loss, and reduced vascular stem cells leading to chronic secondary complications of diabetes.


Assuntos
Adipogenia/efeitos dos fármacos , Glucose/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Antígeno AC133 , Idoso , Angiopoietina-2/metabolismo , Animais , Antígenos CD/metabolismo , Medula Óssea/patologia , Diferenciação Celular/efeitos dos fármacos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Glicoproteínas/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Modelos Biológicos , Peptídeos/metabolismo , Proteína Quinase C/metabolismo , Ratos Sprague-Dawley , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
14.
Pediatr Res ; 75(3): 381-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24296797

RESUMO

BACKGROUND: Infantile hemangioma (IH) is the most common tumor of infancy. The first-line therapy for IH is propranolol, a nonselective ß-adrenergic receptor antagonist. However, mechanisms for the therapeutic effect of propranolol and regrowth of IH following cessation of treatment in some cases are not clear. We have recently shown that IH arises from multipotent stem cells. Whether IH stem cells are responsive to propranolol and are selectively targeted is unknown, and this is the focus of this study. METHODS: IH stem cells were exposed to propranolol and were assayed for cellular and molecular alterations. We used endothelial cells (ECs) as controls and bone marrow-derived mesenchymal progenitor cells (bm-MPCs) as normal stem/progenitor counterparts to determine selectivity. RESULTS: Our results show that propranolol significantly reduced IH stem cell growth but failed to induce caspase-3 activation. Normal bm-MPCs and mature ECs showed maintained or increased caspase-3 activation and significantly reduced cyclin-D1 levels. We further show that IH stem cells may escape apoptosis by inducing antiapoptotic pathways. CONCLUSION: This study reveals that propranolol does not induce apoptosis in IH stem cells, which is in contrast with the result for ECs. Escape from apoptosis in IH stem cells may involve induction of antiapoptotic pathways.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Proliferação de Células/efeitos dos fármacos , Hemangioma/tratamento farmacológico , Células-Tronco Multipotentes/efeitos dos fármacos , Propranolol/farmacologia , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Contagem de Células , Ciclina D1/metabolismo , Hemangioma/etiologia , Humanos , Lactente , Reação em Cadeia da Polimerase em Tempo Real
15.
Adipocyte ; 3(4): 263-72, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-26317050

RESUMO

Diabetes leads to complications in select organ systems primarily by disrupting the vasculature of the target organs. These complications include both micro- (cardiomyopathy, retinopathy, nephropathy, and neuropathy) and macro-(atherosclerosis) angiopathies. Bone marrow angiopathy is also evident in both experimental models of the disease as well as in human diabetes. In addition to vascular disruption, bone loss and increased marrow adiposity have become hallmarks of the diabetic bone phenotype. Emerging evidence now implicates enhanced marrow adipogenesis and changes to cellular makeup of the marrow in a novel mechanistic link between various secondary complications of diabetes. In this review, we explore the mechanisms of enhanced marrow adipogenesis in diabetes and the link between changes to marrow cellular composition, and disruption and depletion of reparative stem cells.

16.
Dermatoendocrinol ; 6(1): e979699, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-26413184

RESUMO

Infantile hemangioma is a common tumor of infancy. Although most hemangiomas spontaneously regress, treatment is indicated based on complications, risk to organ development and function, and disfigurement. The serendipitous discovery of propranolol, a non-selective ß-adrenergic receptor blocker, as an effective means to regress hemangiomas has made this a first-line therapy for hemangioma patients. Propranolol has shown remarkable response rates. There are, however, some adverse effects, which include changes in sleep, acrocyanosis, hypotension, and hypoglycemia. Over the last few years, researchers have focused on understanding the mechanisms by which propranolol causes hemangioma regression. This has entailed study of cultured vascular endothelial cells including endothelial cells isolated from hemangioma patients. In this article, we review recent studies offering potential mechanisms of how various cell types found in hemangioma may respond to propranolol.

17.
Dermatoendocrinol ; 5(3): 352-7, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24516689

RESUMO

Infantile hemangioma is a benign vascular tumor that affects 4 to 10% of neonates. A unique feature of hemangiomas is the natural lifecycle, whereby the tumor rapidly grows and then spontaneously regresses to a fibrofatty residuum. We have shown that hemangiomas are derived from mutlipotential stem cells (hemSCs), which differentiate into endothelial cells during the early proliferating phase and into adipocytes during the later involutive phase. T-box 2 (TBX2) is a transcription factor involved in controlling cell-fate decisions, and is highly expressed during the proliferating phase of hemangioma development. We hypothesize that TBX2 expression would be high in hemSCs derived from human hemangiomas and inhibiting TBX2 would result in changes in hemSC differentiation potential. To test our hypothesis, we analyzed hemSCs for TBX2 mRNA and protein expression. We then used RNA interference and TBX2 overexpression to determine the effect of altering TBX2 levels on hemSC growth and differentiation. Our studies show that TBX2 is highly expressed in hemSCs compared with a panel of normal stem/progenitor cells and mature vascular cells. TBX2 knockdown completely abolished adipogenic differentiation of hemSCs without significantly altering growth. Furthermore, overexpression of TBX2 led to enhanced adipogenic differentiation ability possibly through induction of C/EBPß. From these findings, we believe that TBX2 is active in hemSCs and that TBX2 maintains adipogenic differentiation-competent state of hemSCs. These findings may be important in the development of better treatment options for hemangiomas to accelerate involution.

18.
Exp Mol Pathol ; 94(1): 126-36, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23047069

RESUMO

Infantile hemangioma is a benign vascular tumor that exhibits a unique yet predictable lifecycle of rapid proliferation followed by spontaneous regression. Recent studies have identified that insulin-like growth factor-2 (IGF2), a fetal mitogen, is highly expressed during the proliferative phase of hemangioma growth. Since hemangiomas arise from CD133+ stem cells, high levels of IGF2 may regulate the activity of the stem cells and therefore, hemangioma growth. The aim of this study was to understand the functional significance of elevated IGF2 in hemangiomas. We show that IGF2 localizes to the CD133+ cells in hemangioma specimens. We, therefore, hypothesized that IGF2 may be regulating the plasticity of hemangioma stem cells. To test our hypothesis, we used CD133-selected cells from hemangiomas to knockdown the expression of IGF2. We found that IGF2 is a mitogen for hemangioma stem cells and prevents leptin induction and full terminal differentiation of hemangioma stem cells into adipocytes. We also show that IGF2 does not alter the initial commitment phase. These findings implicate an important role of IGF2 in expanding hemangioma stem cells and preventing terminal adipocyte differentiation.


Assuntos
Adipócitos/fisiologia , Adipogenia , Hemangioma/metabolismo , Hemangioma/patologia , Fator de Crescimento Insulin-Like II/metabolismo , Leptina/metabolismo , Células-Tronco Neoplásicas/fisiologia , Antígeno AC133 , Adipócitos/metabolismo , Antígenos CD/análise , Diferenciação Celular , Proliferação de Células , Glicoproteínas/análise , Hemangioma/irrigação sanguínea , Humanos , Fator de Crescimento Insulin-Like II/genética , Mitógenos , Células-Tronco Neoplásicas/metabolismo , Peptídeos/análise , Interferência de RNA , RNA Interferente Pequeno , Células Tumorais Cultivadas
19.
Invest Ophthalmol Vis Sci ; 53(13): 8333-43, 2012 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-23150625

RESUMO

PURPOSE: Diabetic retinopathy entails proliferation of vascular endothelial cells (ECs) and unregulated angiogenesis. We have previously shown that ECs increase the expression of an embryonic variant of fibronectin (FN), called extra domain-B FN (ED-B FN) in response to high glucose. We also showed that ED-B FN regulates EC tube morphogenesis, possibly through vascular endothelial growth factor (VEGF). In the present study, we have attempted to decipher the mechanisms by which ED-B FN may modulate EC phenotype. METHODS: We hypothesized that ED-B FN regulates VEGF expression in ECs through interaction with selected integrin receptors. To test this hypothesis, we first cultured ECs in high levels of glucose to investigate for any alteration. We then used integrin-specific matrix mimetic peptides, neutralizing antibodies, and RNAi to identify the integrin(s) involved in VEGF expression. Finally, we used an animal model of diabetes to study whether these in vitro mechanisms also take place in the retina. RESULTS: Our results show that exposure of ECs to high levels of glucose increased VEGF expression. ED-B FN mediated this increase since knockdown of ED-B FN completely prevented glucose-induced VEGF expression. We then identified ß1 integrin as the essential receptor involved in high glucose-induced VEGF expression. We also showed that diabetes increased ß1 integrin and VEGF expression in the retina, which normalized upon ED-B knockdown. CONCLUSIONS: These findings showed that high levels of glucose in diabetes increased VEGF expression in ECs through ED-B FN and ß1 integrin interaction. These results provide novel mechanistic basis of increased VEGF expression in diabetes.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Células Endoteliais/efeitos dos fármacos , Fibronectinas/metabolismo , Regulação da Expressão Gênica/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Anticorpos Neutralizantes , Western Blotting , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Células Endoteliais/metabolismo , Fibronectinas/genética , Técnica Indireta de Fluorescência para Anticorpo , Glucose/toxicidade , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Integrina alfa2beta1/antagonistas & inibidores , Integrina alfa2beta1/metabolismo , Integrina alfa5beta1/antagonistas & inibidores , Integrina alfa5beta1/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Vasos Retinianos/citologia , Transfecção
20.
Can J Neurol Sci ; 39(6): 821-7, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23041404

RESUMO

BACKGROUND: Hemangioblastomas are benign vascular tumors of the central nervous system that occur sporadically or in association with von Hippel-Lindau disease. These tumors are characteristically composed of a dense capillary network with intervening stromal/interstitial cells. To date, the histogenesis of hemangioblastoma remains unclear. We hypothesize that hemangioblastomas arise from a defective mesodermal stem cell, which gives rise to the atypical vasculature. METHODS: To test our hypothesis, we have characterized the cellular composition of hemangioblastomas by immunophenotyping pluripotent and committed stem cells and vascular endothelial cells. RESULTS: Our findings show that hemangioblastoma endothelial cells are positive for CD133, a stem and progenitor cell marker. Vascular endothelial cells also expressed nuclear Oct4. In addition to the endothelium, both CD133 and Oct4 were present in stromal and perivascular cells. Interestingly, neither the endothelium nor the stromal cells expressed Sox2 or Nanog suggesting a committed stem cell phenotype. CONCLUSIONS: From these findings, we believe that hemangioblastoma stromal cells are committed stem cells producing both vascular cell types. The findings also show an unusual CD133-positive endothelial phenotype in hemangioblastoma.


Assuntos
Neoplasias Cerebelares/patologia , Hemangioblastoma/patologia , Células-Tronco/fisiologia , Células Estromais/fisiologia , Antígeno AC133 , Actinas/metabolismo , Adulto , Antígenos CD/metabolismo , Diferenciação Celular/fisiologia , Feminino , Transportador de Glucose Tipo 1/metabolismo , Glicoproteínas/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Pessoa de Meia-Idade , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/metabolismo , Peptídeos/metabolismo , Fosfopiruvato Hidratase/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Fatores de Transcrição SOXB1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...