Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros













Intervalo de año de publicación
1.
Biochem Pharmacol ; 223: 116177, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38552853

RESUMEN

Nowadays, the treatment of musculoskeletal diseases represents a major challenge in the developed world. Diseases such as osteoporosis, osteoarthritis and arthritis have a high incidence and prevalence as a consequence of population aging, and they are also associated with a socioeconomic burden. Many efforts have been made to find a treatment for these diseases with various levels of success, but new approaches are still needed to deal with these pathologies. In this context, one peptide derived for the C-terminal extreme of the Parathormone related Peptide (PTHrP) called Osteostatin can be useful to treat musculoskeletal diseases. This pentapeptide (TRSAW) has demonstrated both in different in vitro and in vivo models, its role as a molecule with anti-resorptive, anabolic, anti-inflammatory, and anti-antioxidant properties. Our aim with this work is to review the Osteostatin main features, the knowledge of its mechanisms of action as well as its possible use for the treatment of osteoporosis, bone regeneration and fractures and against arthritis given its anti-inflammatory properties.


Asunto(s)
Artritis , Osteoporosis , Fragmentos de Péptidos , Humanos , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Osteoporosis/tratamiento farmacológico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico
2.
Pharmaceutics ; 15(2)2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36839981

RESUMEN

Osteoporosis is the most common type of bone disease. Conventional treatments are based on the use of antiresorptive drugs and/or anabolic agents. However, these treatments have certain limitations, such as a lack of bioavailability or toxicity in non-specific tissues. In this regard, pleiotrophin (PTN) is a protein with potent mitogenic, angiogenic, and chemotactic activity, with implications in tissue repair. On the other hand, mesoporous silica nanoparticles (MSNs) have proven to be an effective inorganic drug-delivery system for biomedical applications. In addition, the surface anchoring of cationic polymers, such as polyethylenimine (PEI), allows for greater cell internalization, increasing treatment efficacy. In order to load and release the PTN to improve its effectiveness, MSNs were successfully internalized in MC3T3-E1 mouse pre-osteoblastic cells and human mesenchymal stem cells. PTN-loaded MSNs significantly increased the viability, mineralization, and gene expression of alkaline phosphatase and Runx2 in comparison with the PTN alone in both cell lines, evidencing its positive effect on osteogenesis and osteoblast differentiation. This proof of concept demonstrates that MSN can take up and release PTN, developing a potent osteogenic and differentiating action in vitro in the absence of an osteogenic differentiation-promoting medium, presenting itself as a possible treatment to improve bone-regeneration and osteoporosis scenarios.

3.
J Cell Physiol ; 237(10): 3927-3943, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35933642

RESUMEN

Osteocytes respond to mechanical forces controlling osteoblast and osteoclast function. Mechanical stimulation decreases osteocyte apoptosis and promotes bone formation. Primary cilia have been described as potential mechanosensors in bone cells. Certain osteogenic responses induced by fluid flow (FF) in vitro are decreased by primary cilia inhibition in MLO-Y4 osteocytes. The parathyroid hormone (PTH) receptor type 1 (PTH1R) modulates osteoblast, osteoclast, and osteocyte effects upon activation by PTH or PTH-related protein (PTHrP) in osteoblastic cells. Moreover, some actions of PTH1R seem to be triggered directly by mechanical stimulation. We hypothesize that PTH1R forms a signaling complex in the primary cilium that is essential for mechanotransduction in osteocytes and affects osteocyte-osteoclast communication. MLO-Y4 osteocytes were stimulated by FF or PTHrP (1-37). PTH1R and primary cilia signaling were abrogated using PTH1R or primary cilia specific siRNAs or inhibitors, respectively. Conditioned media obtained from mechanically- or PTHrP-stimulated MLO-Y4 cells inhibited the migration of preosteoclastic cells and osteoclast differentiation. Redistribution of PTH1R along the entire cilium was observed in mechanically stimulated MLO-Y4 osteocytic cells. Preincubation of MLO-Y4 cells with the Gli-1 antagonist, the adenylate cyclase inhibitor (SQ22536), or with the phospholipase C inhibitor (U73122), affected the migration of osteoclast precursors and osteoclastogenesis. Proteomic analysis and neutralizing experiments showed that FF and PTH1R activation control osteoclast function through the modulation of C-X-C Motif Chemokine Ligand 5 (CXCL5) and interleukin-6 (IL-6) secretion in osteocytes. These novel findings indicate that both primary cilium and PTH1R are necessary in osteocytes for proper communication with osteoclasts and show that mechanical stimulation inhibits osteoclast recruitment and differentiation through CXCL5, while PTH1R activation regulate these processes via IL-6.


Asunto(s)
Interleucina-6 , Osteoclastos , Inhibidores de Adenilato Ciclasa/farmacología , Quimiocinas/metabolismo , Cilios/metabolismo , Medios de Cultivo Condicionados/metabolismo , Interleucina-6/metabolismo , Ligandos , Mecanotransducción Celular , Osteoclastos/metabolismo , Osteocitos/metabolismo , Hormona Paratiroidea/metabolismo , Hormona Paratiroidea/farmacología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Proteómica , Ligando RANK/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/genética , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Fosfolipasas de Tipo C/metabolismo
4.
Vitam Horm ; 120: 345-370, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35953116

RESUMEN

Primary cilia are subcellular structures specialized in sensing different stimuli in a diversity of cell types. In bone, the primary cilium is involved in mechanical sensing and transduction of signals that regulate the behavior of mesenchymal osteoprogenitors, osteoblasts and osteocytes. To perform its functions, the primary cilium modulates a plethora of molecules including those stimulated by the parathyroid hormone (PTH) receptor type I (PTH1R), a master regulator of osteogenesis. Binding of the agonists PTH or PTH-related protein (PTHrP) to the PTH1R or direct agonist-independent stimulation of the receptor activate PTH1R signaling pathways. In turn, activation of PTH1R leads to regulation of bone formation and remodeling. Herein, we describe the structure, function and molecular partners of primary cilia in the context of bone, playing special attention to those signaling pathways that are mediated directly or indirectly by PTH1R in association with primary cilia during the process of osteogenesis.


Asunto(s)
Osteogénesis , Hormona Paratiroidea , Cilios/metabolismo , Humanos , Osteoblastos , Hormona Paratiroidea/química , Hormona Paratiroidea/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo
5.
Curr Osteoporos Rep ; 18(6): 796-802, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33184775

RESUMEN

PURPOSE OF REVIEW: Diabetes mellitus is a prevalent chronic disease affecting millions of people in the world. Bone fragility is a complication found in diabetic patients. Although osteoblasts and osteoclasts are directly affected by diabetes, herein we focus on how the diabetic state-based on hyperglycemia and accumulation of advanced glycation end products among other features-impairs osteocyte functions exerting deleterious effects on bone. RECENT FINDINGS: In the last years, several studies described that diabetic conditions cause morphological modifications on lacunar-canalicular system, alterations on osteocyte mechanoreceptors and intracellular pathways and on osteocyte communication with other cells through the secretion of proteins such as sclerostin or RANKL. This article gives an overview of events occurring in diabetic osteocytes. In particular, mechanical responses seem to be seriously affected in these conditions, suggesting that mechanical sensibility could be a target for future research in the field.


Asunto(s)
Diabetes Mellitus/fisiopatología , Osteocitos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Fenómenos Biomecánicos , Densidad Ósea , Diabetes Mellitus/metabolismo , Diabetes Mellitus Experimental/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo
6.
Endocr Relat Cancer ; 27(7): 441-456, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32357310

RESUMEN

Bone metastases are common in advanced prostate cancer patients, but mechanisms by which specific pro-metastatic skeletal niches are formed before tumor cell homing are unclear. We aimed to analyze the effects of proteins secreted by primary prostate tumors on the bone microenvironment before the settlement and propagation of metastases. Here, using an in vivo pre-metastatic prostate cancer model based on the implantation of prostate adenocarcinoma TRAMP-C1 cells in immunocompetent C57BL/6 mice, we identify MINDIN as a prostate tumor secreted protein that induces bone microstructural and bone remodeling gene expression changes before tumor cell homing. Associated with these changes, increased tumor cell adhesion to the endosteum ex vivo and to osteoblasts in vitro was observed. Furthermore, MINDIN promoted osteoblast proliferation and mineralization and monocyte expression of osteoclast markers. ß-catenin signaling pathway revealed to mediate MINDIN actions on osteoblast gene expression but failed to affect MINDIN-induced adhesion to prostate tumor cells or monocyte differentiation to osteoclasts. Our study evidences that MINDIN secretion by primary prostate tumors creates a favorable bone environment for tumor cell homing before metastatic spread.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , beta Catenina/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Transfección
7.
Science ; 368(6497): 1371-1376, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32439659

RESUMEN

The effect of immunometabolism on age-associated diseases remains uncertain. In this work, we show that T cells with dysfunctional mitochondria owing to mitochondrial transcription factor A (TFAM) deficiency act as accelerators of senescence. In mice, these cells instigate multiple aging-related features, including metabolic, cognitive, physical, and cardiovascular alterations, which together result in premature death. T cell metabolic failure induces the accumulation of circulating cytokines, which resembles the chronic inflammation that is characteristic of aging ("inflammaging"). This cytokine storm itself acts as a systemic inducer of senescence. Blocking tumor necrosis factor-α signaling or preventing senescence with nicotinamide adenine dinucleotide precursors partially rescues premature aging in mice with Tfam-deficient T cells. Thus, T cells can regulate organismal fitness and life span, which highlights the importance of tight immunometabolic control in both aging and the onset of age-associated diseases.


Asunto(s)
Envejecimiento Prematuro/inmunología , Proteínas de Unión al ADN/deficiencia , Mitocondrias/metabolismo , Proteínas Mitocondriales/deficiencia , Multimorbilidad , Linfocitos T/metabolismo , Factores de Transcripción/deficiencia , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/prevención & control , Animales , Síndrome de Liberación de Citoquinas/inmunología , Proteínas de Unión al ADN/genética , Femenino , Eliminación de Gen , Inflamación/genética , Inflamación/inmunología , Longevidad , Masculino , Ratones , Ratones Mutantes , Proteínas Mitocondriales/genética , NAD/administración & dosificación , NAD/farmacología , Aptitud Física , Linfocitos T/ultraestructura , Factores de Transcripción/genética , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
8.
J Cell Physiol ; 235(10): 7356-7369, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32039485

RESUMEN

Mechanical stimulation of primary cilia in osteocytes and osteoblasts has been proposed as a mechanism that participates in bone cell survival and skeletal remodeling. Among different signaling pathways stimulated by primary cilia, the hedgehog signaling pathway has been associated with the regulation of bone development. Parathyroid hormone (PTH)-related protein (PTHrP) signaling through PTH 1 receptor (PTH1R) also regulates bone cell survival and remodeling and has been associated with the hedgehog pathway during skeletal development. We hypothesize that primary cilia and PTH1R concomitantly regulate bone remodeling and cell survival and aim to describe the mechanisms that mediate these effects in osteocytes and osteoblasts. Colocalization of PTH1R with primary cilia was observed in control and PTHrP-stimulated MLO-Y4 osteocytic and MC3T3-E1 osteoblastic cells. Activation of PTH1R by PTHrP increased cell survival, osteoblast gene expression (osteocalcin, runt-related transcription factor 2, and bone alkaline phosphatase) and the expression of the hedgehog transcription factor Gli-1 in osteocytes and osteoblasts. These effects were abrogated by small interfering RNAs for the primary cilia protein IFT88 or by a primary cilia specific inhibitor (chloral hydrate). Preincubation of MLO-Y4 osteocytic and MC3T3-E1 osteoblastic cells with the Gli-1 antagonist GANT61 inhibited PTHrP prosurvival actions but did not affect PTHrP-induced overexpression of osteogenic genes. Mechanical stimulation by fluid flow increased the number of primary cilia-presenting cells in osteocytes and osteoblasts. We propose that PTH1R activation induces prosurvival actions via primary cilia- and Gli-1-dependent mechanism and modulates osteogenic responses via a primary cilia-dependent and Gli-1-independent pathway in osteocytes and osteoblasts.


Asunto(s)
Cilios/metabolismo , Osteocitos/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Células 3T3 , Animales , Desarrollo Óseo/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Ratones , Osteoblastos/metabolismo , Osteogénesis/fisiología , Transducción de Señal
9.
Knee ; 27(1): 51-60, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31926672

RESUMEN

OBJECTIVE: Increasing evidence has shown the involvement of the innate immune system and inflammatory response in osteoarthritis (OA) pathogenesis; however, anterior cruciate ligament (ACL) tears are recognized risk factors for development of post-traumatic OA. We investigated (1) whether inflammatory mediators involved in OA pathogenesis are also present at significant concentrations in the knee joint sometime after ACL complete tear and may be considered as prognostic biomarkers of progression to secondary OA; and (2) whether quantification in serum may surrogate synovial fluid (SF) measurements in both cases. METHODS: Thirty-seven end-stage OA patients and 33 patients with ACL complete tear that were included on the waiting list for knee surgery were consecutively recruited. Serum and SF samples were taken before surgery, and tumor necrosis factor-alpha, (TNF-α), interleukin-6 (IL-6), interleukin-8 (IL-8), matrix metalloproteinase-1 (MMP1), matrix metalloproteinase-3 (MMP3), tissue inhibitor of metalloproteinase-1 (TIMP1), bone morphogenetic protein-7 (BMP7), regulated upon activation normal t-cell expressed and secreted (RANTES), cytokine interferon-γ-induced protein 10 (IP-10) and heat shock protein family A (Hsp70) member 1A (HSPA1A) were quantified by enzyme-linked immunosorbent assay (ELISA.) Normally distributed data were compared using a one-way analysis of variance (ANOVA) test. Data not normally distributed were analyzed using a nonparametric Mann-Whitney rank sum test. Differences in means were compared using a Student's t-test. Correlations were analyzed using Pearson's coefficient of variation. RESULTS: Eighty-seven percent of patients with OA and 86% of those with ACL tear had quantifiable levels of biomarkers in SF. SF levels of IL-6, IL-8, MMP1, MMP3, RANTES, IP-10, BMP7 and HSPA1A were significantly lower in ACL injury knees compared with those with OA, but much higher than those reported in control subjects. Serum levels of IL-6, IP-10, and MMP1 were also lower in patients with ACL tears, who had, however, significantly higher TNF-α, HSPA1A, and TIMP1 levels when compared with OA patients. Levels of biomarkers tested in serum and SF samples were significantly different. CONCLUSIONS: Our data propose that cytokines IL-6 and IL-8 and the chemokines RANTES, IP-10, MMP1, MMP3, and HSPA1A may be involved in the inflammatory process leading to synovitis, the central lesion in OA onset and development; persistent high levels of these substances sometime after ACL injury suggest that they could play an etiopathogenic role in the maintenance of the inflammatory environment leading to post-traumatic OA. Serum biomarker levels do not appear to faithfully reflect what occurs inside the joint. Thus, most biomarkers cannot yet be considered as useful inflammatory biomarkers of knee joint diseases.


Asunto(s)
Lesiones del Ligamento Cruzado Anterior/metabolismo , Ligamento Cruzado Anterior/cirugía , Citocinas/metabolismo , Osteoartritis de la Rodilla/metabolismo , Líquido Sinovial/metabolismo , Adulto , Anciano , Lesiones del Ligamento Cruzado Anterior/complicaciones , Lesiones del Ligamento Cruzado Anterior/cirugía , Reconstrucción del Ligamento Cruzado Anterior/métodos , Biomarcadores/metabolismo , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Osteoartritis de la Rodilla/etiología
10.
Front Pharmacol ; 10: 1255, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31708781

RESUMEN

Macrophages occupy a prominent position during immune responses. They are considered the final effectors of any given immune response since they can be activated by a wide range of surface ligands and cytokines to acquire a continuum of functional states. Macrophages are involved in tissue homeostasis and in the promotion or resolution of inflammatory responses, causing tissue damage or helping in tissue repair. Knowledge in macrophage polarization has significantly increased in the last decade. Biomarkers, functions, and metabolic states associated with macrophage polarization status have been defined both in murine and human models. Moreover, a large body of evidence demonstrated that macrophage status is a dynamic process that can be modified. Macrophages orchestrate virtually all major diseases-sepsis, infection, chronic inflammatory diseases (rheumatoid arthritis), neurodegenerative disease, and cancer-and thus they represent attractive therapeutic targets. In fact, the possibility to "reprogram" macrophage status is considered as a promising strategy for designing novel therapies. Here, we will review the role of different tissue macrophage populations in the instauration and progression of inflammatory and non-inflammatory pathologies, as exemplified by rheumatoid arthritis, osteoporosis, glioblastoma, and tumor metastasis. We will analyze: 1) the potential as therapeutic targets of recently described macrophage populations, such as osteomacs, reported to play an important role in bone formation and homeostasis or metastasis-associated macrophages (MAMs), key players in the generation of premetastatic niche; 2) the current and potential future approaches to target monocytes/macrophages and their inflammation-causing products in rheumatoid arthritis; and 3) the development of novel intervention strategies using oncolytic viruses, immunomodulatory agents, and checkpoint inhibitors aiming to boost M1-associated anti-tumor immunity. In this review, we will focus on the potential of macrophages as therapeutic targets and discuss their involvement in state-of-the-art strategies to modulate prevalent pathologies of aging societies.

11.
Trends Endocrinol Metab ; 30(10): 756-766, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31409530

RESUMEN

Musculoskeletal disorders represent an elevated socioeconomic burden for developed aging societies. Osteoporosis (OP) has been treated with antiresorptive therapies or with teriparatide that was until recently the only anabolic therapy. However, approval of osteoporosis treatment in postmenopausal women with abaloparatide, which is an analog of parathyroid hormone-related peptide (PTHrP), has created a new alternative for OP management. The success of this new treatment is related to differential mechanisms of activation of PTH receptor type 1 (PTH1R) by abaloparatide and PTH. Here, we address the distinguishing mechanisms of PTH1R activation; the effects of PTH1R stimulation in osteoblast, osteocytes, and chondrocytes; the differences between PTH and abaloparatide actions on PTH1R; potential safety concerns; and future perspectives about abaloparatide use in other musculoskeletal disorders.


Asunto(s)
Proteína Relacionada con la Hormona Paratiroidea/uso terapéutico , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteocitos/efectos de los fármacos , Osteocitos/metabolismo , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Osteoporosis Posmenopáusica/tratamiento farmacológico , Osteoporosis Posmenopáusica/metabolismo , Teriparatido/uso terapéutico
12.
J Cell Physiol ; 234(10): 18917-18927, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30912165

RESUMEN

The infrapatellar fat pad (IPFP) is a periarticular adipose knee tissue. This tissue contains a large number of mesenchymal stem cells (MSCs). In the present work, we wanted to study the IPFP MSCs and their relationship and differences in two groups, anterior cruciate ligament (ACL) ruptures knees and ostheoarthrosis (OA). The IPFP of 42 patients with OA or ACL rupture were analyzed. Isolation, primary culture, and a genetic and proteomic study of MSCs from IPFP were performed. Gene expression of IL-6, tumor necrosis factor (TNF), IL-8, HSPA1A (Hsp70), CXCL10, RANTES, MMP1, MMP3, TIMP1, and BMP7 was analyzed by real-time quantitative polymerase chain reaction (RT-qPCR). We analyzed MSCs from from 12 diferents patients in two cellular pools (6 from AO disease and 6 from ALC rupture to form two cell pool), for the iTRAQ Proteomic Assay. The conditional media were used in quantitative analysis of MSC soluble factors by Luminex and for de migration assay. A higher gene expression of IL-6, TNF, CXCL10, RANTES, and MMP1 and OPG in MSCs from OA versus ACL (p < 0.05) was observed. Conversely HSPA1A, TIMP1, and RANKL showed a significant lower expression in OA-MSCs (p < 0.05). In the secretome analysis, adipsin and visfantin levels in the supernatants from OA-MSCs were lower (p < 0.05) respect to ACL-MSCs. Also, the monocytic cells migrated two-folds in the presence of conditioned media from OA-MSCs patients versus patients with ACL-MSC. The infrapatellar pad should be considered as an adipose tissue capable of producing and excreting inflammatory mediators directly in the knee joint, influencing the development and progression of knee joint pathologies.


Asunto(s)
Tejido Adiposo/metabolismo , Lesiones del Ligamento Cruzado Anterior/patología , Articulación de la Rodilla/citología , Células Madre Mesenquimatosas/metabolismo , Osteoartritis/patología , Rótula/citología , Tejido Adiposo/citología , Adulto , Ligamento Cruzado Anterior/citología , Ligamento Cruzado Anterior/patología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Transcriptoma , Adulto Joven
13.
Sci Rep ; 9(1): 2295, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30783155

RESUMEN

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.


Asunto(s)
Ácido Cítrico/metabolismo , Glucosa/farmacología , Osteocitos/efectos de los fármacos , Osteocitos/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Cromatografía Liquida , Electroforesis Capilar , Manitol/farmacología , Espectrometría de Masas , Mecanotransducción Celular , Metabolómica , Ratones , Análisis de Componente Principal
14.
Cartilage ; 9(1): 55-62, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29156945

RESUMEN

Aim In the osteoarthritis (OA) disease, all structures of the joint are involved. The infrapatellar Hoffa fat pad is rich in macrophages and granulocytes, which also represents a source of adipose mesenchymal progenitor cells (ASC) cells. In our study, we analyze how OA affects the ability of ASC-derived from Hoffa's fat pad to differentiate into chondrocytes. Material and methodology We took knee Hoffa's pad samples and adipose tissue from the proximal thigh from 6 patients diagnosed with severe OA and from another 6 patients with an anterior cruciate ligament (ACL) rupture without OA. From all the patients, we took subcutaneous adipose tissue from the thigh, as the control group. Samples of synovial fluid (SF) were also extracted. The gene expression was analyzed by real-time quantitative polymerase chain reaction. Results PTH1R and MMP13 expression during chondrogenic differentiation were similar between OA and ACL groups, while the expression of OPG, FGF2, TGFß, MMP3 were significantly lower in the OA group. Exposure of differentiated ASC to OA SF induced an increase in the expression of OPG, PTH1R, and MMP13 and a decrease in the expression of FGF2 in cell culture of the ACL group. However, expression of none of these factors was altered by the OA synovial fluid in ASC cells of the OA group. Conclusion OA of the knee also affects the mesenchymal stem cells of Hoffa fat, suggesting that Hoffa fat is a new actor in the OA degenerative process that can contribute to the origin, onset, and progression of the disease.


Asunto(s)
Articulación de la Rodilla/patología , Células Madre Mesenquimatosas/metabolismo , Osteoartritis de la Rodilla/metabolismo , Grasa Subcutánea/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Lesiones del Ligamento Cruzado Anterior/metabolismo , Condrocitos/metabolismo , Estudios Transversales , Expresión Génica , Humanos , Metaloproteinasa 13 de la Matriz/metabolismo , Persona de Mediana Edad , Ensayos Clínicos Controlados no Aleatorios como Asunto , Osteoartritis de la Rodilla/diagnóstico , Estudios Prospectivos , Líquido Sinovial/metabolismo , Muslo/patología
15.
Aging Cell ; 16(3): 551-563, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28317237

RESUMEN

Skeletal aging results in apoptosis of osteocytes, cells embedded in bone that control the generation/function of bone forming and resorbing cells. Aging also decreases connexin43 (Cx43) expression in bone; and osteocytic Cx43 deletion partially mimics the skeletal phenotype of old mice. Particularly, aging and Cx43 deletion increase osteocyte apoptosis, and osteoclast number and bone resorption on endocortical bone surfaces. We examined herein the molecular signaling events responsible for osteocyte apoptosis and osteoclast recruitment triggered by aging and Cx43 deficiency. Cx43-silenced MLO-Y4 osteocytic (Cx43def ) cells undergo spontaneous cell death in culture through caspase-3 activation and exhibit increased levels of apoptosis-related genes, and only transfection of Cx43 constructs able to form gap junction channels reverses Cx43def cell death. Cx43def cells and bones from old mice exhibit reduced levels of the pro-survival microRNA miR21 and, consistently, increased levels of the miR21 target phosphatase and tensin homolog (PTEN) and reduced phosphorylated Akt, whereas PTEN inhibition reduces Cx43def cell apoptosis. miR21 reduction is sufficient to induce apoptosis of Cx43-expressing cells and miR21 deletion in miR21fl/fl bones increases apoptosis-related gene expression, whereas a miR21 mimic prevents Cx43def cell apoptosis, demonstrating that miR21 lies downstream of Cx43. Cx43def cells release more osteoclastogenic cytokines [receptor activator of NFκB ligand (RANKL)/high-mobility group box-1 (HMGB1)], and caspase-3 inhibition prevents RANKL/HMGB1 release and the increased osteoclastogenesis induced by conditioned media from Cx43def cells, which is blocked by antagonizing HMGB1-RAGE interaction. These findings identify a novel Cx43/miR21/HMGB1/RANKL pathway involved in preventing osteocyte apoptosis that also controls osteoclast formation/recruitment and is impaired with aging.


Asunto(s)
Envejecimiento/metabolismo , Conexina 43/genética , MicroARNs/genética , Osteoclastos/metabolismo , Osteocitos/metabolismo , Osteogénesis/genética , Envejecimiento/patología , Animales , Apoptosis/efectos de los fármacos , Resorción Ósea/genética , Resorción Ósea/metabolismo , Resorción Ósea/patología , Huesos/metabolismo , Huesos/patología , Caspasa 3/genética , Caspasa 3/metabolismo , Conexina 43/deficiencia , Medios de Cultivo Condicionados/farmacología , Femenino , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/metabolismo , Uniones Comunicantes/patología , Regulación de la Expresión Génica , Prueba de Complementación Genética , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Osteocitos/efectos de los fármacos , Osteocitos/patología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Transducción de Señal
16.
J Cell Physiol ; 232(12): 3611-3621, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28138960

RESUMEN

Diabetes mellitus (DM) induces bone deterioration, while mechanical stimulation promotes osteocyte-driven bone formation. We aimed to evaluate the interaction of acute exposure (24 h) to high glucose (HG) with both the pro-survival effect conferred to osteocytic MLO-Y4 cells and osteoblastic MC3T3-E1 cells by mechanical stimulation and the interaction of these cells with osteoclast precursor RAW264.7 cells. We found that 24 h of HG (25 mM) pre-exposure prevented both cell survival and ERK and ß-catenin nuclear translocation upon mechanical stimulation by fluid flow (FF) (10 min) in both MLO-Y4 and MC3T3-E1 cells. However, migration of RAW 264.7 cells was inhibited by MLO-Y4 cell-conditioned medium (CM), but not by MC3T3-E1 cell-CM, with HG or FF. This inhibitory effect was associated with consistent changes in VEGF, RANTES, MIP-1α, MIP-1ß MCP-1, and GM-CSF in MLO-Y4 cell-CM. RAW264.7 proliferation was inhibited by MLO-Y4 CM under static or HG conditions, but it increased by FF-CM with or without HG. In addition, both FF and HG abrogated the capacity of RAW 264.7 cells to differentiate into osteoclasts, but in a different manner. Thus, HG-CM in static condition allowed formation of osteoclast-like cells, which were unable to resorb hydroxyapatite. In contrast, FF-CM prevented osteoclastogenesis even in HG condition. Moreover, HG did not affect basal RANKL or IL-6 secretion or their inhibition induced by FF in MLO-Y4 cells. In conclusion, this in vitro study demonstrates that HG exerts disparate effects on osteocyte mechanotransduction, and provides a novel mechanism by which DM disturbs skeletal metabolism through altered osteocyte-osteoclast communication.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Glucosa/farmacología , Mecanotransducción Celular/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteocitos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Células Madre/efectos de los fármacos , Células 3T3 , Animales , Comunicación Celular/efectos de los fármacos , Citocinas/metabolismo , Ratones , Osteoclastos/metabolismo , Osteocitos/metabolismo , Estimulación Física , Células RAW 264.7 , Células Madre/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Histol Histopathol ; 32(8): 751-760, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27981526

RESUMEN

Bone is one of the most adaptable tissues in the body as it is continuously subjected to load bearing. In fact, mechanical loading is an important regulator of bone mass. The skeleton adjusts to load by changing its mass, shape and microarchitecture, depending on the magnitude of the strain. Mechanical stimulation is necessary for the development of the skeleton, whereas in adults physiological levels of strain help maintain bone mass by reducing bone resorption. On the other hand, an excessive level of strain or bone disuse induces bone loss. Osteocytes are long-lived cells comprising more than 90% of bone cellularity, which are embedded in the bone matrix forming a functional syncytium extending to the bone surface. These cells are considered to be the main bone cells responsible for translating mechanical strain into regulatory signals for osteoblasts and osteoclasts, leading to adapting bone responses to environmental changes. In this review, we discuss the complexity and well-orchestrated events that occur in bone mechanotransduction, focusing on osteocyte viability as an important biological response in this respect. Elucidation of the molecular mechanisms of bone mechanotransduction and the key role of osteocytes is opening new avenues for the treatment of bone loss-related diseases.


Asunto(s)
Huesos/fisiología , Mecanotransducción Celular , Osteocitos/citología , Adenosina Trifosfato/química , Animales , Densidad Ósea , Matriz Ósea , Resorción Ósea , Adhesión Celular , Supervivencia Celular , Citoesqueleto/metabolismo , Humanos , Óxido Nítrico/metabolismo , Osteoblastos/citología , Osteoclastos/citología , Osteocitos/metabolismo , Estrés Mecánico
18.
J Cell Physiol ; 232(8): 2158-2167, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27925206

RESUMEN

Mechanical forces, hypoxia, and oxidative stress contribute to skin renewal, perfusion, and wound healing, but how are they regulating subcutaneous adipose-derived stem cells (ASCs) in the inflammatory microenvironment associated to skin repair and disorders is unknown. In this study, ASCs were isolated from lipoaspirate samples from plastic surgery patients, primary cultured and their differentiation and secretion of a panel of cytokines with pronounced effects on skin repair and angiogenesis were studied under mechanical stimulation by intermittent fluid flow, 1% hypoxia and oxidative stress by glutathione (GSH) depletion with buthionine sulfoximine (BSO) treatment. Mechanical action of fluid flow did not alter mesenchymal phenotype of CD90+ /CD29+ /CD44+ /CD34- /CD106- /CD45- ASCs; however, it remarkably induced ASC secretion of human umbilical vein endothelial cell (HUVEC) migration-stimulating factors. Multiplex Luminex assay further confirmed an increased secretion of VEGF, G-CSF, HGF, Leptin, IL-8, PDGF-BB, Angiopoietin-2, and Follistatin from mechanically-stimulated ASCs via cyclooxygenase-2. Consistent with this mechanism, GSH depletion and hypoxia also increased ASC secretion of VEGF, IL-8, leptin, Angiopoitein-2, and PDGF-BB. However, mechanical action of fluid flow abrogated VEGF and HUVEC migration-stimulating activity from GSH-depleted and hypoxic ASCs. Conversely, GSH depletion and hypoxia abrogated VEGF and HUVEC migration-stimulating activity from mechano-stimulated ASCs. Although mechanical action of fluid flow, hypoxia, and GSH-depletion had independent proangiogenic-stimulating activity on ASCs, mechanical stimulation had opposite effects on proangiogenic factor secretion from ASCs with and without oxidative stress. These data uncover the role of hypoxia and endogenous redox balance during the proangiogenic response of ASCs and other mesenchymal-derived cell types to mechanical action of interstitial fluid flow. J. Cell. Physiol. 232: 2158-2167, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Tejido Adiposo/metabolismo , Proteínas Angiogénicas/metabolismo , Citocinas/metabolismo , Mecanotransducción Celular , Estrés Oxidativo , Células Madre/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Adulto , Butionina Sulfoximina/farmacología , Hipoxia de la Célula , Separación Celular/métodos , Células Cultivadas , Quimiotaxis , Medios de Cultivo Condicionados/metabolismo , Femenino , Glutatión/deficiencia , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Mecanotransducción Celular/efectos de los fármacos , Persona de Mediana Edad , Neovascularización Fisiológica , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Comunicación Paracrina , Fenotipo , Cultivo Primario de Células , Nicho de Células Madre , Células Madre/efectos de los fármacos , Estrés Mecánico
19.
J Bone Miner Res ; 32(3): 486-497, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27683064

RESUMEN

There is an unmet need to understand the mechanisms underlying skeletal deterioration in diabetes mellitus (DM) and to develop therapeutic approaches to treat bone fragility in diabetic patients. We demonstrate herein that mice with type 1 DM induced by streptozotocin exhibited low bone mass, inferior mechanical and material properties, increased bone resorption, decreased bone formation, increased apoptosis of osteocytes, and increased expression of the osteocyte-derived bone formation inhibitor Sost/sclerostin. Further, short treatment of diabetic mice with parathyroid hormone related protein (PTHrP)-derived peptides corrected these changes to levels undistinguishable from non-diabetic mice. In addition, diabetic mice exhibited reduced bone formation in response to mechanical stimulation, which was corrected by treatment with the PTHrP peptides, and higher prevalence of apoptotic osteocytes, which was reduced by loading or by the PTHrP peptides alone and reversed by a combination of loading and PTHrP peptide treatment. In vitro experiments demonstrated that the PTHrP peptides or mechanical stimulation by fluid flow activated the survival kinases ERKs and induced nuclear translocation of the canonical Wnt signaling mediator ß-catenin, and prevented the increase in osteocytic cell apoptosis induced by high glucose. Thus, PTHrP-derived peptides cross-talk with mechanical signaling pathways to reverse skeletal deterioration induced by DM in mice. These findings suggest a crucial role of osteocytes in the harmful effects of diabetes on bone and raise the possibility of targeting these cells as a novel approach to treat skeletal deterioration in diabetes. Moreover, our study suggests the potential therapeutic efficacy of combined pharmacological and mechanical stimuli to promote bone accrual and maintenance in diabetic subjects. © 2016 American Society for Bone and Mineral Research.


Asunto(s)
Huesos/anatomía & histología , Huesos/fisiología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Proteínas Adaptadoras Transductoras de Señales , Adiposidad/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Fenómenos Biomecánicos , Densidad Ósea/efectos de los fármacos , Resorción Ósea/genética , Resorción Ósea/patología , Huesos/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/fisiopatología , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Tamaño de los Órganos/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteocitos/efectos de los fármacos , Osteocitos/metabolismo , Osteogénesis/efectos de los fármacos , Soporte de Peso
20.
Actual. osteol ; 12(1): 11-20, 2016. ilus
Artículo en Inglés | LILACS, UNISALUD, BINACIS | ID: biblio-1379682

RESUMEN

Bisphosphonates (BPs) anti-fracture efficacy may be due in part to inhibition of osteocyte apoptosis. This effect requires opening of connexin (Cx) 43 hemichannels and phosphorylation of the extracellular signal regulated kinases (ERKs). However, unlike ERK activation by other stimuli, the Cx43/ERK pathway activated by BPs does not result in nuclear ERK accumulation. Instead, the anti-apoptotic effect of BPs depends on phosphorylation of cytoplasmic ERK targets and is abolished by forced nuclear retention of ERKs. We now report that ERKs and the scaffolding protein ß-arrestin co-immuno-precipitate with Cx43 in MLO-Y4 osteocytic cells and that the BP alendronate increases this association. Moreover, ERK2 fused to red fluorescent protein (ERK2-RFP) co-localizes with Cx43 fused to green fluorescent protein outside the nucleus in cells untreated or treated with alendronate. Alendronate does not induce ERK nuclear accumulation in cells transfected with wild type ß-arrestin (wtARR) or vector control, whereas it does in cells expressing a dominant negative ß-arrestin mutant (dnARR) consisting of the ß-arrestin-clathrin binding domain that competes with endogenous ß-arrestin for binding to clathrin. Alendronate activates ERKs in dnARRtransfected cells as effectively as in cells transfected with wtARR, demonstrating that dnARR only interferes with subcellular localization but not with activation of ERKs by BPs. Further, whereas alendronate inhibits apoptosis in cells expressing wtARR or vector control, it is ineffective in cells expressing dnARR. Thus, BPs induce the formation of a complex comprising Cx43, ß-arrestin, and clathrin, which directs ERKs outside the nucleus and is indispensable for osteocyte survival induced by BPs. (AU)


La efectividad de los bisfosfonatos (BPs) en la prevención de fracturas puede deberse en parte a la inhibición de la apoptosis de osteocitos. Este efecto depende de la apertura de hemicanales de conexina (Cx) 43 y la fosforilación de quinasas reguladas por señales extracelulares (ERKs). Sin embargo, a diferencia de la activación de ERKs debida a otros estímulos, la vía de señalización Cx43/ERK activada por BPs no conlleva la acumulación de ERKs en el núcleo. El efecto anti-apoptótico de los BPs depende de la fosforilación de blancos citoplasmáticos de ERKs y es inhibido cuando las quinasas son retenidas en el núcleo. En este estudio hemos demostrado que ERKs y la proteína "scaffolding" ß-arrestina co-inmunoprecipitan con Cx43 en células osteocíticas MLO-Y4 y que alendronato aumenta esta asociación. Más aún, ERK2 fusionada a la proteína roja fluorescente (ERK2-RFP) co-localiza con Cx43 fusionada con la proteína verde fluorescente fuera del núcleo en células tratadas con vehículo o alendronato. Alendronato no indujo la acumulación nuclear de ERK en células transfectadas con ß-arrestina nativa (wtARR) o con un vector control, pero si lo hizo en células que expresan una forma dominante negativa de ß-arrestina (dnARR), consistente en el dominio de interacción entre ß-arrestina y clatrina, y que compite con ß-arrestina endógena por la unión a clatrina. Alendronato activa ERKs con la misma eficiencia en células transfectadas con dnARR o wtARR, demostrando que dnARR sólo interfiere con la localización subcelular de ERKs, pero no con su activación inducida por los BPs. Más aún, mientras alendronato inhibe apoptosis en células que expresan wtARR o vector control, es inefectivo en células que expresan dnARR. En conclusión, los BPs inducen la formación de un complejo que incluye Cx43, ß-arrestina y clatrina, el cual retiene ERKs fuera del núcleo y es indispensable para la sobrevida de los osteocitos inducida por estas drogas. (AU)


Asunto(s)
Osteocitos/citología , Núcleo Celular/enzimología , Apoptosis/efectos de los fármacos , Conexina 43/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Difosfonatos/farmacología , beta-Arrestinas/metabolismo , Osteocitos/efectos de los fármacos , Osteocitos/metabolismo , Huesos/citología , Supervivencia Celular/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA