Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Rev. osteoporos. metab. miner. (Internet) ; 15(1): 12-20, Ene-Mar. 2023. graf, ilus
Artigo em Espanhol | IBECS | ID: ibc-218430

RESUMO

Introducción: los osteocitos son capaces de detectar diferentes señales, transducirlas en respuestas biológicas y trasmitirlasa los osteoblastos y osteoclastos, permitiendo el mantenimiento de la homeostasis ósea. La mecanotransducción ósea esposible gracias a que los osteocitos presentan diferentes estructuras mecanosensoras como las conexinas (Cx), las integrinas,el cilio primario e incluso receptores acoplados a proteínas G como el receptor de la parathormona tipo 1 (PTH1R).Objetivo: analizar la posible interacción de los diferentes elementos mecanosensores de los osteocitos y ver su influen-cia en la respuesta biológica.Material y métodos: se trabajó con las líneas celulares osteocíticas MLO-Y4 Cx43+/+ (scrambled (SCR) y ARNi α2) yCx43-/-.Resultados y conclusión: los resultados obtenidos muestran que la Cx43 y la integrina α2 se encuentran involucradas enel aumento de la longitud del cilio primario, afectando potencialmente a su funcionalidad como mecanosensor (SCR vs.ARNi α2, p < 0,0001 SCR vs. Cx43-/- y p < 0,0001 ARNi α2 vs. Cx43-/-). La integrina α2 también influyó en la localizacióncelular de Cx43 promoviendo que esta se encuentre en la membrana plasmática. También se observó que la activación dePTH1R por agonistas como parathormona (PTH) y proteína relacionada con la parathormona (PTHrP) inducen la fosforilaciónde la quinasa ERK 1/2, y estos efectos podrían verse afectados por la deficiencia en Cx43, pero no parecen ser mediadospor el silenciamiento de integrina α2. Finalmente, se observó que la presencia de la Cx43 y de integrina α2 en los osteoci-tos aumenta su capacidad de adhesión (Cx43+/+ SCR y ARNi α2 vs. CX43-/- p < 0,001 y p = 0,0039) y que la deficienciaen Cx43 provoca un incremento de la mortalidad de estas células (Cx43-/- vs. Cx43+/+ p = 0,0074).(AU)


Assuntos
Humanos , Distúrbios do Metabolismo do Cálcio , Osteoblastos , Cílios , Conexinas , Integrinas , Osteócitos , Doenças Ósseas , Osteoporose
2.
Rev. osteoporos. metab. miner. (Internet) ; 15(1): 21-28, Ene-Mar. 2023. graf, ilus
Artigo em Espanhol | IBECS | ID: ibc-218431

RESUMO

El esqueleto es un órgano metabólicamente activo que se remodela continuamente a la largo de nuestra vida. Estaremodelación implica un equilibrio entre la formación de hueso llevada a cabo por los osteoblastos y la resorción porlos osteoclastos. Los osteocitos son los encargados de regular estos dos procesos y su estimulación mecánica, es esen-cial para mantener el buen funcionamiento óseo y prevenir enfermedades como la osteoporosis. La estimulación de lososteocitos provoca una alteración en la producción y secreción de moléculas de señalización que regulan la actividadde los osteoblastos y los osteoclastos. Las células madre mesenquimales han sido propuestas como posible terapiacelular para la regeneración de distintos tejidos, incluido el tejido óseo. Hipotetizamos en el presente estudio que elsecretoma de células osteocíticas de ratón estimuladas mecánicamente afecta a la capacidad proliferativa, adhesiva ya la expresión génica de células mesenquimales indiferenciadas y células mesenquimales preosteoblásticas. Para ello,se analizaron los procesos biológicos mencionados en líneas continuas celulares preosteoblásticas y células mesenqui-males de ratón en presencia del medio condicionado por células osteocíticas MLO-Y4, después de ser sometidas aestímulo mecánico por flujo de fluido. Se observó que la proliferación aumentó en ambas líneas celulares en presenciadel secretoma de osteocitos estimulados mecánicamente frente al control, mientras que osteocitos no mecanoestimu-lados provocaban su disminución. También se observó un aumento en la capacidad adhesiva de células C3H/10T1/2 trasla estimulación con el secretoma de osteocitos mecanoestimulados. En cuanto a la expresión de genes, solo el factoradipogénico PPARᵞ sufrió alteraciones en células MC3T3-E1 por el secretoma de osteocitos. Estos estudios indican quelos osteocitos pueden modificar el comportamiento biológico de células mesenquimales mediante la secreción de factores solubles.(AU)


Assuntos
Humanos , Osteócitos , Esqueleto , Células-Tronco Mesenquimais , Osteoblastos , Osteoclastos , Reabsorção Óssea , Osteoporose , Doenças Ósseas
3.
Sci Rep ; 9(1): 2295, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30783155

RESUMO

Osteocytes are the main cells of bone tissue and play a crucial role in bone formation and resorption. Recent studies have indicated that Diabetes Mellitus (DM) affects bone mass and potentially causes higher bone fracture risk. Previous work on osteocyte cell cultures has demonstrated that mechanotransduction is impaired after culture under diabetic pre-conditioning with high glucose (HG), specifically osteoclast recruitment and differentiation. The aim of this study was to analyze the extracellular metabolic changes of osteocytes regarding two conditions: pre-conditioning to either basal levels of glucose (B), mannitol (M) or HG cell media, and mechanical stimulation by fluid flow (FF) in contrast to static condition (SC). Secretomes were analyzed using Liquid Chromatography and Capillary Electrophoresis both coupled to Mass Spectrometry (LC-MS and CE-MS, respectively). Results showed the osteocyte profile was very similar under SC, regardless of their pre-conditioning treatment, while, after FF stimulation, secretomes followed different metabolic signatures depending on the pre-conditioning treatment. An important increment of citrate pointed out that osteocytes release citrate outside of the cell to induce osteoblast activation, while HG environment impaired FF effect. This study demonstrates for the first time that osteocytes increase citrate excretion under mechanical stimulation, and that HG environment impaired this effect.


Assuntos
Ácido Cítrico/metabolismo , Glucose/farmacologia , Osteócitos/efeitos dos fármacos , Osteócitos/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cromatografia Líquida , Eletroforese Capilar , Manitol/farmacologia , Espectrometria de Massas , Mecanotransdução Celular , Metabolômica , Camundongos , Análise de Componente Principal
4.
Oncotarget ; 8(11): 18456-18485, 2017 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-28060743

RESUMO

Worldwide deaths from diabetes mellitus (DM) and colorectal cancer increased by 90% and 57%, respectively, over the past 20 years. The risk of colorectal cancer was estimated to be 27% higher in patients with type 2 DM than in non-diabetic controls. However, there are potential confounders, information from lower income countries is scarce, across the globe there is no correlation between DM prevalence and colorectal cancer incidence and the association has evolved over time, suggesting the impact of additional environmental factors. The clinical relevance of these associations depends on understanding the mechanism involved. Although evidence is limited, insulin use has been associated with increased and metformin with decreased incidence of colorectal cancer. In addition, colorectal cancer shares some cellular and molecular pathways with diabetes target organ damage, exemplified by diabetic kidney disease. These include epithelial cell injury, activation of inflammation and Wnt/ß-catenin pathways and iron homeostasis defects, among others. Indeed, some drugs have undergone clinical trials for both cancer and diabetic kidney disease. Genome-wide association studies have identified diabetes-associated genes (e.g. TCF7L2) that may also contribute to colorectal cancer. We review the epidemiological evidence, potential pathophysiological mechanisms and therapeutic implications of the association between DM and colorectal cancer. Further studies should clarify the worldwide association between DM and colorectal cancer, strengthen the biological plausibility of a cause-and-effect relationship through characterization of the molecular pathways involved, search for specific molecular signatures of colorectal cancer under diabetic conditions, and eventually explore DM-specific strategies to prevent or treat colorectal cancer.


Assuntos
Neoplasias Colorretais/epidemiologia , Diabetes Mellitus Tipo 2/epidemiologia , Antineoplásicos/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Humanos , Hipoglicemiantes/uso terapêutico
5.
J Gerontol A Biol Sci Med Sci ; 71(3): 290-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26386012

RESUMO

In the present study, the possibility that a diabetic (DM) status might worsen age-related bone deterioration was explored in mice. Male CD-1 mice aged 2 (young control group) or 16 months, nondiabetic or made diabetic by streptozotocin injections, were used. DM induced a decrease in bone volume, trabecular number, and eroded surface, and in mineral apposition and bone formation rates, but an increased trabecular separation, in L1-L3 vertebrae of aged mice. Three-point bending and reference point indentation tests showed slight changes pointing to increased frailty and brittleness in the mouse tibia of diabetic old mice. DM was related to a decreased expression of both vascular endothelial growth factor and its receptor 2, which paralleled that of femoral vasculature, and increased expression of the pro-adipogenic gene peroxisome proliferator-activated receptor γ and adipocyte number, without affecting ß-catenin pathway in old mouse bone. Concomitant DM in old mice failed to affect total glutathione levels or activity of main anti-oxidative stress enzymes, although xanthine oxidase was slightly increased, in the bone marrow, but increased the senescence marker caveolin-1 gene. In conclusion, DM worsens bone alterations of aged mice, related to decreased bone turnover and bone vasculature and increased senescence, independently of the anti-oxidative stress machinery.


Assuntos
Envelhecimento , Densidade Óssea , Diabetes Mellitus Experimental/metabolismo , Osteogênese/fisiologia , Osteoporose/metabolismo , Animais , Remodelação Óssea , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/patologia , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/metabolismo , Vértebras Lombares/patologia , Masculino , Camundongos , Osteoporose/diagnóstico , Osteoporose/etiologia , Tomografia Computadorizada por Raios X
6.
Acta Biomater ; 10(7): 3307-16, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24704694

RESUMO

Biopolymer-coated nanocrystalline hydroxyapatite (HA) made as macroporous foams which are degradable and flexible are promising candidates as orthopaedic implants. The C-terminal (107-111) epitope of parathyroid hormone-related protein (PTHrP) exhibits osteogenic properties. The main aim of this study was to evaluate whether PTHrP (107-111) loading into gelatin-glutaraldehyde biopolymer-coated HA (HAGlu) scaffolds would produce an optimal biomaterial for tissue engineering applications. HAGlu scaffolds with and without PTHrP (107-111) were implanted into a cavitary defect performed in both distal tibial metaphysis of adult rats. Animals were sacrificed after 4 weeks for histological, microcomputerized tomography and gene expression analysis of the callus. At this time, bone healing occurred only in the presence of PTHrP (107-111)-containing HAGlu implant, related to an increase in bone volume/tissue volume and trabecular thickness, cortical thickness and gene expression of osteocalcin and vascular cell adhesion molecule 1, but a decreased gene expression of Wnt inhibitors, SOST and dickkopf homolog 1. The autonomous osteogenic effect of the PTHrP (107-111)-loaded HAGlu scaffolds was confirmed in mouse and human osteoblastic cell cultures. Our findings demonstrate the advantage of loading PTHrP (107-111) into degradable HAGlu scaffolds for achieving an optimal biomaterial that is promising for low load bearing clinical applications.


Assuntos
Biopolímeros/química , Regeneração Óssea/efeitos dos fármacos , Materiais Revestidos Biocompatíveis , Durapatita/química , Gelatina/química , Glutaral/química , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/farmacologia , Células 3T3 , Animais , Sequência de Bases , Primers do DNA , Camundongos , Microscopia Eletrônica de Varredura , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Alicerces Teciduais
7.
Histol Histopathol ; 28(11): 1473-81, 2013 11.
Artigo em Inglês | MEDLINE | ID: mdl-23673876

RESUMO

Oxidative stress in bone increases with age, which leads to bone frailty and a high fracture risk. Animal models show that early changes in trabecular structure occur in age-related osteopenia. These models might be valuable to assess the contribution of oxidative stress in age-related bone loss. Premature aging mice (PAM) have previously been characterized as a model of premature immunological and neurological senescence. PAM long bones (mainly consisting of cortical bone) display features of aging bone. Thus, we aimed to evaluate the vertebrae, representing a unique poorly loaded type of trabecular bone in mice, in PAM and no PAM (NPAM) controls. PAM showed an anxious behaviour, based on physical activity evaluation. These mice had decreased bone mineral density (0.078 mg/cm² in NPAM vs 0.070 g/cm² in PAM; p⟨0.05); a decreased number of osteocytes per bone field (404±36 in NPAM vs 320±27 in PAM; p⟨0.01); and downregulation of various osteoblastic genes and low eroded surface/bone surface, 4.2±0.5 in NPAM vs 1.9±0.2 in PAM; p⟨0.01). This was associated with increased expression of oxidative stress markers, Foxo1 and GADD45, in PAM vertebrae. Mesenchymal progenitors in the bone marrow of PAM have a poor mineralization capacity (assessed by the number of mineralized nodules and suface), and showed a lower response to an osteogenic input -represented by parathormone-related protein-, compared to NPAM. Collectively, these results indicate that PAM vertebrae show osteopenia related to diminished bone formation and remodeling. Our findings further support the validity of PAM as a suitable model for involutional osteoporosis and its treatment.


Assuntos
Envelhecimento/patologia , Modelos Animais de Doenças , Vértebras Lombares/patologia , Osteoporose/patologia , Animais , Feminino , Vértebras Lombares/fisiopatologia , Camundongos , Osteoporose/fisiopatologia , Reação em Cadeia da Polimerase em Tempo Real
8.
J Cell Physiol ; 227(5): 1980-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-21732369

RESUMO

Hypertrophy of human mesangial cells (HMC) is among the earliest characteristics in patients with diabetic nephropathy (DN). Recently, we observed the upregulation of parathyroid hormone (PTH)-related protein (PTHrP) in experimental DN, associated with renal hypertrophy. Herein, we first examined whether PTHrP was overexpressed in human DN, and next assessed the putative role of this protein on high glucose (HG)-induced HMC hypertrophy. As previously found in mice, kidneys from diabetic patients showed an increased tubular and glomerular immunostaining for PTHrP. In HMC, HG medium increased PTHrP protein expression associated with the development of hypertrophy as assessed by cell protein content. This effect was also induced by PTHrP(1-36). HG and PTHrP(1-36)-induced hypertrophy were associated with an increase in cyclin D1 and p27Kip1 protein expression, a decreased cyclin E expression, and the prevention of cyclin E/cdk2 complex activation. Both PTHrP neutralizing antiserum (α-PTHrP) and the PTH/PTHrP receptor antagonist (JB4250) were able to abolish HG induction of hypertrophy, the aforementioned changes in cell cycle proteins, and also TGF-ß1 up-regulation. Moreover, the capability of both HG and PTHrP(1-36) to induce HMC hypertrophy was abolished by α-TGFß1. These data show for the first time that PTHrP is upregulated in the kidney of patients with DN. Our findings also demonstrate that PTHrP acts as an important mediator of HG-induced HMC hypertrophy by modulating cell cycle regulatory proteins and TGF-ß1.


Assuntos
Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Rim/metabolismo , Rim/patologia , Células Mesangiais/metabolismo , Células Mesangiais/patologia , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina E/genética , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Feminino , Humanos , Hipertrofia/metabolismo , Hipertrofia/patologia , Rim/citologia , Masculino , Células Mesangiais/citologia , Camundongos , Pessoa de Meia-Idade , Proteína Relacionada ao Hormônio Paratireóideo/genética , Fator de Crescimento Transformador beta1/metabolismo
9.
J Am Soc Nephrol ; 21(2): 237-48, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19959711

RESUMO

Epithelial-mesenchymal transition (EMT) is an important process that contributes to renal fibrogenesis. TGF-beta1 and EGF stimulate EMT. Recent studies suggested that parathyroid hormone-related protein (PTHrP) promotes fibrogenesis in the damaged kidney, apparently dependent on its interaction with vascular endothelial growth factor (VEGF), but whether it also interacts with TGF-beta and EGF to modulate EMT is unknown. Here, PTHrP(1-36) increased TGF-beta1 in cultured tubuloepithelial cells and TGF-beta blockade inhibited PTHrP-induced EMT-related changes, including upregulation of alpha-smooth muscle actin and integrin-linked kinase, nuclear translocation of Snail, and downregulation of E-cadherin and zonula occludens-1. PTHrP(1-36) also induced EGF receptor (EGFR) activation; inhibition of protein kinase C and metalloproteases abrogated this activation. Inhibition of EGFR activation abolished these EMT-related changes, the activation of ERK1/2, and upregulation of TGF-beta1 and VEGF by PTHrP(1-36). Moreover, inhibition of ERK1/2 blocked EMT induced by either PTHrP(1-36), TGF-beta1, EGF, or VEGF. In vivo, obstruction of mouse kidneys led to changes consistent with EMT and upregulation of TGF-beta1 mRNA, p-EGFR protein, and PTHrP. Taken together, these data suggest that PTHrP, TGF-beta, EGF, and VEGF might cooperate through activation of ERK1/2 to induce EMT in renal tubuloepithelial cells.


Assuntos
Diferenciação Celular , Células Epiteliais/patologia , Túbulos Renais/patologia , Rim/patologia , Mesoderma/patologia , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Fator de Crescimento Epidérmico/metabolismo , Células Epiteliais/metabolismo , Receptores ErbB/metabolismo , Fibrose , Humanos , Rim/metabolismo , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
J Am Soc Nephrol ; 17(6): 1594-603, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16672315

RESUMO

Parathyroid hormone-related protein (PTHrP) is shortly upregulated in acute renal injury, but its pathophysiologic role is unclear. Investigated was whether PTHrP might act as a profibrogenic factor in mice that do or do not overexpress PTHrP in the proximal tubule after folic acid (FA) nephrotoxicity, a model of acute renal damage followed by partial regeneration and patchy tubulointerstitial fibrosis. It was found that constitutive PTHrP overexpression in these animals conveyed a significant increase in tubulointerstitial fibrosis, associated with both fibroblast activation (as alpha-smooth muscle actin staining) and macrophage influx, compared with control littermates at 2 to 3 wk after FA damage. Cell proliferation and survival was higher (P<0.01) in the renal interstitium of PTHrP-overexpressing mice than in control littermates within this period after injury. Moreover, the former mice had a constitutive Bcl-XL protein overexpression. In vitro studies in renal tubulointerstitial and fibroblastic cells strongly suggest that PTHrP (1-36) (100 nM) reduced FA-induced apoptosis through a dual mechanism involving Bcl-XL upregulation and Akt and Bad phosphorylation. PTHrP (1-36) also stimulated monocyte chemoattractant protein-1 expression in tubuloepithelial cells, as well as type-1 procollagen gene expression and fibronectin (mRNA levels and protein secretion) in these cells and renal fibroblastic cells. Our findings indicate that this peptide, by interaction with the PTH1 receptor, can increase tubulointerstitial cell survival and seems to act as a proinflammatory and profibrogenic factor in the FA-damaged kidney.


Assuntos
Apoptose , Ácido Fólico/toxicidade , Túbulos Renais/efeitos dos fármacos , Rim/efeitos dos fármacos , Proteína Relacionada ao Hormônio Paratireóideo/fisiologia , Actinas/metabolismo , Animais , Proliferação de Células , Matriz Extracelular/metabolismo , Fibrose , Rim/lesões , Túbulos Renais/lesões , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Liso/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...