Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Vasc Res ; 58(5): 277-285, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33951626

RESUMEN

The sodium-dependent phosphate transporter, SLC20A1, is required for elevated inorganic phosphate (Pi) induced vascular smooth muscle cell (VSMC) matrix mineralization and phenotype transdifferentiation. Recently, elevated Pi was shown to induce ERK1/2 phosphorylation through SLC20A1 by Pi uptake-independent functions in VSMCs, suggesting a cell signaling response to elevated Pi. Previous studies identified Rap1 guanine nucleotide exchange factor (RapGEF1) as an SLC20A1-interacting protein and RapGEF1 promotes ERK1/2 phosphorylation through Rap1 activation. In this study, we tested the hypothesis that RapGEF1 is a critical component of the SLC20A1-mediated Pi-induced ERK1/2 phosphorylation pathway. Co-localization of SLC20A1 and RapGEF1, knockdown of RapGEF1 with siRNA, and small molecule inhibitors of Rap1, B-Raf, and Mek1/2 were investigated. SLC20A1 and RapGEF1 were co-localized in peri-membranous structures in VSMCs. Knockdown of RapGEF1 and small molecule inhibitors against Rap1, B-Raf, and Mek1/2 eliminated elevated Pi-induced ERK1/2 phosphorylation. Knockdown of RapGEF1 inhibited SM22α mRNA expression and blocked elevated Pi-induced downregulation of SM22α mRNA. Together, these data suggest that RapGEF1 is required for SLC20A1-mediated elevated Pi signaling through a Rap1/B-Raf/Mek1/2 cell signaling pathway, thereby promoting ERK1/2 phosphorylation and inhibiting SM22α gene expression in VSMCs.


Asunto(s)
Factor 2 Liberador de Guanina Nucleótido/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Fosfatos/farmacología , Animales , Células Cultivadas , Factor 2 Liberador de Guanina Nucleótido/genética , Humanos , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Fosforilación , Transducción de Señal , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo
2.
Kidney Int ; 94(4): 716-727, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30041812

RESUMEN

PiT-2, a type III sodium-dependent phosphate transporter, is a causative gene for the brain arteriolar calcification in people with familial basal ganglion calcification. Here we examined the effect of PiT-2 haploinsufficiency on vascular calcification in uremic mice using wild-type and global PiT-2 heterozygous knockout mice. PiT-2 haploinsufficiency enhanced the development of vascular calcification in mice with chronic kidney disease fed a high-phosphate diet. No differences were observed in the serum mineral biomarkers and kidney function between the wild-type and PiT-2 heterozygous knockout groups. Micro computed tomography analyses of femurs showed that haploinsufficiency of PiT-2 decreased trabecular bone mineral density in uremia. In vitro, sodium-dependent phosphate uptake was decreased in cultured vascular smooth muscle cells isolated from PiT-2 heterozygous knockout mice compared with those from wild-type mice. PiT-2 haploinsufficiency increased phosphate-induced calcification of cultured vascular smooth muscle cells compared to the wild-type. Furthermore, compared to wild-type vascular smooth muscle cells, PiT-2 deficient vascular smooth muscle cells had lower osteoprotegerin levels and increased matrix calcification, which was attenuated by osteoprotegerin supplementation. Thus, PiT-2 in vascular smooth muscle cells protects against phosphate-induced vascular calcification and may be a therapeutic target in the chronic kidney disease population.


Asunto(s)
Fosfatos/metabolismo , Insuficiencia Renal Crónica/complicaciones , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Calcificación Vascular/genética , Animales , Biomarcadores/sangre , Densidad Ósea/genética , Femenino , Haploinsuficiencia , Heterocigoto , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/metabolismo , Osteoprotegerina/metabolismo , Fosfatos/administración & dosificación , Insuficiencia Renal Crónica/sangre , Uremia/complicaciones , Calcificación Vascular/sangre
3.
Biochem Biophys Res Commun ; 495(1): 553-559, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29133259

RESUMEN

Normal bone mineralization requires phosphate oversaturation in bone matrix vesicles, as well as normal regulation of phosphate metabolism via the interplay among bone, intestine, and kidney. In turn, derangement of phosphate metabolism greatly affects bone function and structure. The type III sodium-dependent phosphate transporters, PiT-1 and PiT-2, are believed to be important in tissue phosphate metabolism and physiological bone formation, but their requirement and molecular roles in bone remain poorly investigated. In order to decipher the role of PiT-2 in bone, we examined normal bone development, growth, and mineralization in global PiT-2 homozygous knockout mice. PiT-2 deficiency resulted in reduced vertebral column, femur, and tibia length as well as mandibular dimensions. Micro-computed tomography analysis revealed that bone mineral density in the mandible, femur, and tibia were decreased, indicating that maintenance of bone function and structure is impaired in both craniofacial and long bones of PiT-2 deficient mice. Both cortical and trabecular thickness and mineral density were reduced in PiT-2 homozygous knockout mice compared with wild-type mice. These results suggest that PiT-2 is involved in normal bone development and growth and plays roles in cortical and trabecular bone metabolism feasibly by regulating local phosphate transport and mineralization processes in the bone. Further studies that evaluate bone cell-specific loss of PiT-2 are now warranted and may yield insight into complex mechanisms of bone development and growth, leading to identification of new therapeutic options for patients with bone diseases.


Asunto(s)
Densidad Ósea , Desarrollo Óseo , Enfermedades Óseas/metabolismo , Enfermedades Óseas/patología , Huesos/patología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética
4.
Kidney Int ; 89(5): 1027-1036, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27083280

RESUMEN

Pathologic calcification is a significant cause of increased morbidity and mortality in patients with chronic kidney disease. The precise mechanisms of ectopic calcification are not fully elucidated, but it is known to be caused by an imbalance of procalcific and anticalcific factors. In the chronic kidney disease population, an elevated phosphate burden is both highly prevalent and a known risk factor for ectopic calcification. Here we tested whether osteopontin, an inhibitor of calcification, protects against high phosphate load-induced nephrocalcinosis and vascular calcification. Osteopontin knockout mice were placed on a high phosphate diet for 11 weeks. Osteopontin deficiency together with phosphate overload caused uremia, nephrocalcinosis characterized by substantial renal tubular and interstitial calcium deposition, and marked vascular calcification when compared with control mice. Although the osteopontin-deficient mice did not exhibit hypercalcemia or hyperphosphatemia, they did show abnormalities in the mineral metabolism hormone fibroblast growth factor-23. Thus, endogenous osteopontin plays a critical role in the prevention of phosphate-induced nephrocalcinosis and vascular calcification in response to high phosphate load. A better understanding of osteopontin's role in phosphate-induced calcification will hopefully lead to better biomarkers and therapies for this disease, especially in patients with chronic kidney disease and other at-risk populations.


Asunto(s)
Aorta Abdominal/metabolismo , Aorta Torácica/metabolismo , Enfermedades de la Aorta/prevención & control , Riñón/metabolismo , Nefrocalcinosis/prevención & control , Osteopontina/metabolismo , Fosfatos , Calcificación Vascular/prevención & control , Animales , Aorta Abdominal/patología , Aorta Torácica/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Biomarcadores/sangre , Remodelación Ósea , Dieta , Modelos Animales de Enfermedad , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Riñón/patología , Ratones Endogámicos DBA , Ratones Noqueados , Nefrocalcinosis/genética , Nefrocalcinosis/metabolismo , Nefrocalcinosis/patología , Osteopontina/deficiencia , Osteopontina/genética , Calcificación Vascular/genética , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
5.
Am J Pathol ; 185(7): 1958-69, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25987250

RESUMEN

Arterial medial calcification (AMC) is a hallmark of aging, diabetes, and chronic kidney disease. Smooth muscle cell (SMC) transition to an osteogenic phenotype is a common feature of AMC, and is preceded by expression of runt-related transcription factor 2 (Runx2), a master regulator of bone development. Whether SMC-specific Runx2 expression is required for osteogenic phenotype change and AMC remains unknown. We therefore created an improved targeting construct to generate mice with floxed Runx2 alleles (Runx2(f/f)) that do not produce truncated Runx2 proteins after Cre recombination, thereby preventing potential off-target effects. SMC-specific deletion using SM22-recombinase transgenic allele mice (Runx2(ΔSM)) led to viable mice with normal bone and arterial morphology. After vitamin D overload, arterial SMCs in Runx2(f/f) mice expressed Runx2, underwent osteogenic phenotype change, and developed severe AMC. In contrast, vitamin D-treated Runx2(ΔSM) mice had no Runx2 in blood vessels, maintained SMC phenotype, and did not develop AMC. Runx2 deletion did not affect serum calcium, phosphate, fibroblast growth factor-23, or alkaline phosphatase levels. In vitro, Runx2(f/f) SMCs calcified to a much greater extent than those derived from Runx2(ΔSM) mice. These data indicate a critical role of Runx2 in SMC osteogenic phenotype change and mineral deposition in a mouse model of AMC, suggesting that Runx2 and downstream osteogenic pathways in SMCs may be useful therapeutic targets for treating or preventing AMC in high-risk patients.


Asunto(s)
Calcio/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Miocitos del Músculo Liso/metabolismo , Calcificación Vascular/metabolismo , Animales , Desarrollo Óseo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Miocitos del Músculo Liso/patología , Osteogénesis/efectos de los fármacos , Fenotipo , Fosfatos/metabolismo , Eliminación de Secuencia , Calcificación Vascular/patología , Vitamina D/efectos adversos
6.
Exp Cell Res ; 333(1): 39-48, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25684711

RESUMEN

Vascular calcification (VC) is prevalent in chronic kidney disease and elevated serum inorganic phosphate (Pi) is a recognized risk factor. The type III sodium-dependent phosphate transporter, PiT-1, is required for elevated Pi-induced osteochondrogenic differentiation and matrix mineralization in vascular smooth muscle cells (VSMCs). However, the molecular mechanism(s) by which PiT-1 promotes these processes is unclear. In the present study, we confirmed that the Pi concentration required to induce osteochondrogenic differentiation and matrix mineralization of mouse VSMCs was well above that required for maximal Pi uptake, suggesting a signaling function of PiT-1 that was independent of Pi transport. Elevated Pi-induced signaling via ERK1/2 phosphorylation was abrogated in PiT-1 deficient VSMCs, but could be rescued by wild-type (WT) and a Pi transport-deficient PiT-1 mutant. Furthermore, both WT and transport-deficient PiT-1 mutants promoted osteochondrogenic differentiation as measured by decreased SM22α and increased osteopontin mRNA expression. Finally, compared to vector alone, expression of transport-deficient PiT-1 mutants promoted VSMC matrix mineralization, but not to the extent observed with PiT-1 WT. These data suggest that both Pi uptake-dependent and -independent functions of PiT-1 are important for VSMC processes mediating vascular calcification.


Asunto(s)
Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/fisiología , Animales , Transporte Biológico , Diferenciación Celular , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfatos/metabolismo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Calcificación Vascular/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 34(11): 2387-93, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25189570

RESUMEN

Calcific aortic valve disease (CAVD) is increasingly prevalent worldwide with significant morbidity and mortality. Therapeutic options beyond surgical valve replacement are currently limited. In 2011, the National Heart Lung and Blood Institute assembled a working group on aortic stenosis. This group identified CAVD as an actively regulated disease process in need of further study. As a result, the Alliance of Investigators on CAVD was formed to coordinate and promote CAVD research, with the goals of identifying individuals at risk, developing new therapeutic approaches, and improving diagnostic methods. The group is composed of cardiologists, geneticists, imaging specialists, and basic science researchers. This report reviews the current status of CAVD research and treatment strategies with identification of areas in need of additional investigation for optimal management of this patient population.


Asunto(s)
Estenosis de la Válvula Aórtica/terapia , Válvula Aórtica/patología , Investigación Biomédica/tendencias , Calcinosis/terapia , Cardiopatías Congénitas/terapia , Enfermedades de las Válvulas Cardíacas/terapia , Válvula Aórtica/fisiopatología , Estenosis de la Válvula Aórtica/diagnóstico , Estenosis de la Válvula Aórtica/fisiopatología , Enfermedad de la Válvula Aórtica Bicúspide , Calcinosis/diagnóstico , Calcinosis/fisiopatología , Procedimientos Quirúrgicos Cardíacos , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/fisiopatología , Enfermedades de las Válvulas Cardíacas/diagnóstico , Enfermedades de las Válvulas Cardíacas/fisiopatología , Implantación de Prótesis de Válvulas Cardíacas , Hemodinámica/fisiología , Humanos , Transducción de Señal/fisiología
8.
Am J Physiol Renal Physiol ; 307(8): F891-900, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25143458

RESUMEN

Patients with chronic kidney disease (CKD) and end-stage renal disease (ESRD) have significant cardiovascular morbidity and mortality that is in part due to the development of vascular calcification. Vascular calcification is an active, highly regulated process that shares many similarities with normal bone formation. New discoveries related to extracellular vesicles, microRNAs, and calciprotein particles continue to reveal the mechanisms that are involved in the initiation and progression of vascular calcification in CKD. Further innovations in these fields are critical for the development of biomarkers and therapeutic options for patients with CKD and ESRD.


Asunto(s)
Fallo Renal Crónico/fisiopatología , Insuficiencia Renal Crónica/fisiopatología , Calcificación Vascular/fisiopatología , Calcio/metabolismo , Progresión de la Enfermedad , Elastina/metabolismo , Humanos , Músculo Liso Vascular/citología , Fósforo/metabolismo , Factores de Riesgo , Calcificación Vascular/patología
9.
Arterioscler Thromb Vasc Biol ; 33(11): 2625-32, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23968976

RESUMEN

OBJECTIVE: Elevated serum phosphate has emerged as a major risk factor for vascular calcification. The sodium-dependent phosphate cotransporter, PiT-1, was previously shown to be required for phosphate-induced osteogenic differentiation and calcification of cultured human vascular smooth muscle cells (VSMCs), but its importance in vascular calcification in vivo and the potential role of its homologue, PiT-2, have not been determined. We investigated the in vivo requirement for PiT-1 in vascular calcification using a mouse model of chronic kidney disease and the potential compensatory role of PiT-2 using in vitro knockdown and overexpression strategies. APPROACH AND RESULTS: Mice with targeted deletion of PiT-1 in VSMCs were generated (PiT-1(Δsm)). PiT-1 mRNA levels were undetectable, whereas PiT-2 mRNA levels were increased 2-fold in the vascular aortic media of PiT-1(Δsm) compared with PiT-1(flox/flox) control. When arterial medial calcification was induced in PiT-1(Δsm) and PiT-1(flox/flox) by chronic kidney disease followed by dietary phosphate loading, the degree of aortic calcification was not different between genotypes, suggesting compensation by PiT-2. Consistent with this possibility, VSMCs isolated from PiT-1(Δsm) mice had no PiT-1 mRNA expression, increased PiT-2 mRNA levels, and no difference in sodium-dependent phosphate uptake or phosphate-induced matrix calcification compared with PiT-1(flox/flox) VSMCs. Knockdown of PiT-2 decreased phosphate uptake and phosphate-induced calcification of PiT-1(Δsm) VSMCs. Furthermore, overexpression of PiT-2 restored these parameters in human PiT-1-deficient VSMCs. CONCLUSIONS: PiT-2 can mediate phosphate uptake and calcification of VSMCs in the absence of PiT-1. Mechanistically, PiT-1 and PiT-2 seem to serve redundant roles in phosphate-induced calcification of VSMCs.


Asunto(s)
Músculo Liso Vascular/metabolismo , Insuficiencia Renal Crónica/fisiopatología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Calcificación Vascular/fisiopatología , Animales , Aorta/citología , Aorta/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Fosfatos/metabolismo , ARN Mensajero/metabolismo , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Uremia/genética , Uremia/metabolismo , Uremia/fisiopatología , Calcificación Vascular/genética , Calcificación Vascular/metabolismo
10.
Nucleic Acids Res ; 40(11): e85, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22402491

RESUMEN

The main methods for producing genetically engineered cells use viral vectors for which safety issues and manufacturing costs remain a concern. In addition, selection of desired cells typically relies on the use of cytotoxic drugs with long culture times. Here, we introduce an efficient non-viral approach combining the Sleeping Beauty (SB) Transposon System with selective proliferation of engineered cells by chemically induced dimerization (CID) of growth factor receptors. Minicircles carrying a SB transposon cassette containing a reporter transgene and a gene for the F36VFGFR1 fusion protein were delivered to the hematopoietic cell line Ba/F3. Stably-transduced Ba/F3 cell populations with >98% purity were obtained within 1 week using this positive selection strategy. Copy number analysis by quantitative PCR (qPCR) revealed that CID-selected cells contain on average higher copy numbers of transgenes than flow cytometry-selected cells, demonstrating selective advantage for cells with multiple transposon insertions. A diverse population of cells is present both before and after culture in CID media, although site-specific qPCR of transposon junctions show that population diversity is significantly reduced after selection due to preferential expansion of clones with multiple integration events. This non-viral, positive selection approach is an attractive alternative for producing engineered cells.


Asunto(s)
Ingeniería Celular/métodos , Elementos Transponibles de ADN , Animales , Línea Celular , Línea Celular Transformada , Proliferación Celular , Supervivencia Celular , Dimerización , Ingeniería Genética/métodos , Proteínas Fluorescentes Verdes/genética , Ratones , Plásmidos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Tacrolimus/análogos & derivados , Tacrolimus/metabolismo , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo , Transducción Genética , Transgenes
11.
J Vis Exp ; (205)2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38587386

RESUMEN

This protocol details the propagation and passaging of human iPSCs and their differentiation into osteoclasts. First, iPSCs are dissociated into a single-cell suspension for further use in embryoid body induction. Following mesodermal induction, embryoid bodies undergo hematopoietic differentiation, producing a floating hematopoietic cell population. Subsequently, the harvested hematopoietic cells undergo a macrophage colony-stimulating factor maturation step and, finally, osteoclast differentiation. After osteoclast differentiation, osteoclasts are characterized by staining for TRAP in conjunction with a methyl green nuclear stain. Osteoclasts are observed as multinucleated, TRAP+ polykaryons. Their identification can be further supported by Cathepsin K staining. Bone and mineral resorption assays allow for functional characterization, confirming the identity of bona fide osteoclasts. This protocol demonstrates a robust and versatile method to differentiate human osteoclasts from iPSCs and allows for easy adoption in applications requiring large quantities of functional human osteoclasts. Applications in the areas of bone research, cancer research, tissue engineering, and endoprosthesis research could be envisioned.


Asunto(s)
Resorción Ósea , Células Madre Pluripotentes Inducidas , Humanos , Osteoclastos , Diferenciación Celular , Factor Estimulante de Colonias de Macrófagos/farmacología , Huesos , Glicoproteínas de Membrana , Ligando RANK
12.
J Cell Biochem ; 114(5): 1194-202, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23192608

RESUMEN

Osteopontin (OPN) is highly expressed by macrophages and plays a key role in the pathology of several chronic inflammatory diseases including atherosclerosis and the foreign body reaction. However, the molecular mechanism behind OPN regulation of macrophage functions is not well understood. OPN is a secreted molecule and interacts with several integrins via two domains: the RGD sequence binding to α(v) -containing integrins, and the SLAYGLR sequence binding to α(4) ß(1), α(4) ß(7), and α(9) ß(1) integrins. Here we determined the role of OPN in macrophage survival, chemotaxis, and activation state. For survival studies, OPN treated-bone marrow derived macrophages (BMDMs) were challenged with growth factor withdrawal and neutralizing integrin antibodies. We found that survival in BMDMs is mediated primarily through the α(4) integrin. In chemotaxis studies, we observed that migration to OPN was blocked by neutralizing α(4) and α(9) integrin antibodies. Further, OPN did not affect macrophage activation as measured by IL-12 production. Finally, the relative contributions of the RGD and the SLAYGLR functional domains of OPN to leukocyte recruitment were evaluated in an in vivo model. We generated chimeric mice expressing mutated forms of OPN in myeloid-derived leukocytes, and found that the SLAYGLR functional domain of OPN, but not the RGD, mediates macrophage accumulation in response to thioglycollate-elicited peritonitis. Collectively, these data indicate that α(4) and α(9) integrins interacting with OPN via the SLAYGLR domain play a key role in macrophage biology by regulating migration, survival, and accumulation.


Asunto(s)
Quimiotaxis , Cadenas alfa de Integrinas/metabolismo , Integrina alfa4/metabolismo , Macrófagos/citología , Osteopontina/metabolismo , Secuencia de Aminoácidos , Animales , Células de la Médula Ósea/citología , Supervivencia Celular , Citocinas/biosíntesis , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Osteopontina/química , Peritonitis/metabolismo , Peritonitis/patología , Fenotipo , Estructura Terciaria de Proteína , Transducción de Señal , Relación Estructura-Actividad , Tioglicolatos
13.
Kidney Int ; 83(6): 1159-68, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23389416

RESUMEN

Elevated fibroblast growth factor 23 (FGF23) is associated with cardiovascular disease in patients with chronic kidney disease. As a potential mediating mechanism, FGF23 induces left ventricular hypertrophy; however, its role in arterial calcification is less clear. In order to study this, we quantified coronary artery and thoracic aorta calcium by computed tomography in 1501 patients from the Chronic Renal Insufficiency Cohort (CRIC) study within a median of 376 days (interquartile range 331-420 days) of baseline. Baseline plasma FGF23 was not associated with the prevalence or severity of coronary artery calcium after multivariable adjustment. In contrast, higher serum phosphate levels were associated with prevalence and severity of coronary artery calcium, even after adjustment for FGF23. Neither FGF23 nor serum phosphate were consistently associated with thoracic aorta calcium. We could not detect mRNA expression of FGF23 or its coreceptor, klotho, in human or mouse vascular smooth muscle cells, or normal or calcified mouse aorta. Whereas elevated phosphate concentrations induced calcification in vitro, FGF23 had no effect on phosphate uptake or phosphate-induced calcification regardless of phosphate concentration or even in the presence of soluble klotho. Thus, in contrast to serum phosphate, FGF23 is not associated with arterial calcification and does not promote calcification experimentally. Hence, phosphate and FGF23 promote cardiovascular disease through distinct mechanisms.


Asunto(s)
Aorta Torácica/metabolismo , Enfermedades de la Aorta/sangre , Calcio/metabolismo , Enfermedad de la Arteria Coronaria/sangre , Vasos Coronarios/metabolismo , Factores de Crecimiento de Fibroblastos/sangre , Insuficiencia Renal Crónica/sangre , Calcificación Vascular/sangre , Adulto , Anciano , Animales , Aorta Torácica/diagnóstico por imagen , Enfermedades de la Aorta/diagnóstico por imagen , Enfermedades de la Aorta/epidemiología , Aortografía/métodos , Células Cultivadas , Distribución de Chi-Cuadrado , Angiografía Coronaria/métodos , Enfermedad de la Arteria Coronaria/diagnóstico por imagen , Enfermedad de la Arteria Coronaria/epidemiología , Vasos Coronarios/diagnóstico por imagen , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Glucuronidasa/genética , Glucuronidasa/metabolismo , Humanos , Proteínas Klotho , Modelos Logísticos , Masculino , Ratones , Persona de Mediana Edad , Análisis Multivariante , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Fosfatos/sangre , Prevalencia , Estudios Prospectivos , ARN Mensajero/metabolismo , Insuficiencia Renal Crónica/diagnóstico por imagen , Insuficiencia Renal Crónica/epidemiología , Factores de Riesgo , Índice de Severidad de la Enfermedad , Factores de Tiempo , Tomografía Computarizada por Rayos X , Estados Unidos/epidemiología , Regulación hacia Arriba , Calcificación Vascular/diagnóstico por imagen , Calcificación Vascular/epidemiología , Adulto Joven
14.
Calcif Tissue Int ; 93(4): 365-73, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23456027

RESUMEN

Vascular calcification is highly associated with cardiovascular disease mortality, particularly in high-risk patients with diabetes and chronic kidney diseases (CKD). In blood vessels, intimal calcification is associated with atherosclerosis, whereas medial calcification is a nonocclusive process which leads to increased vascular stiffness and reduced vascular compliance. In the valves, calcification of the leaflets can change the mechanical properties of the tissue and result in stenosis. For many decades, vascular calcification has been noted as a consequence of aging. Studies now confirm that vascular calcification is an actively regulated process and shares many features with bone development and metabolism. This review provides an update on the mechanisms of vascular calcification including the emerging roles of the RANK/RANKL/OPG triad, osteoclasts, and microRNAs. Potential treatments adapted from osteoporosis and CKD treatments that are under investigation for preventing and/or regressing vascular calcification are also reviewed.


Asunto(s)
Calcinosis/metabolismo , Calcificación Vascular/metabolismo , Calcificación Vascular/terapia , Animales , Anticuerpos Monoclonales Humanizados/química , Aterosclerosis/patología , Calcinosis/terapia , Calcio/metabolismo , Quelantes/química , Denosumab , Difosfonatos/química , Humanos , Ratones , MicroARNs/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Osteoporosis/terapia , Osteoprotegerina/metabolismo , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/terapia , Teriparatido/química , Túnica Íntima/patología , Túnica Media/patología , Enfermedades Vasculares/complicaciones
15.
Circ Res ; 109(6): 697-711, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21885837

RESUMEN

Vascular calcification contributes to the high risk of cardiovascular mortality in chronic kidney disease (CKD) patients. Dysregulation of calcium (Ca) and phosphate (P) metabolism is common in CKD patients and drives vascular calcification. In this article, we review the physiological regulatory mechanisms for Ca and P homeostasis and the basis for their dysregulation in CKD. In addition, we highlight recent findings indicating that elevated Ca and P have direct effects on vascular smooth muscle cells (VSMCs) that promote vascular calcification, including stimulation of osteogenic/chondrogenic differentiation, vesicle release, apoptosis, loss of inhibitors, and extracellular matrix degradation. These studies suggest a major role for elevated P in promoting osteogenic/chondrogenic differentiation of VSMC, whereas elevated Ca has a predominant role in promoting VSMC apoptosis and vesicle release. Furthermore, the effects of elevated Ca and P are synergistic, providing a major stimulus for vascular calcification in CKD. Unraveling the complex regulatory pathways that mediate the effects of both Ca and P on VSMCs will ultimately provide novel targets and therapies to limit the destructive effects of vascular calcification in CKD patients.


Asunto(s)
Calcinosis/sangre , Calcio/fisiología , Enfermedades Cardiovasculares/sangre , Fallo Renal Crónico/sangre , Fosfatos/fisiología , Animales , Arterias/metabolismo , Arterias/patología , Calcinosis/patología , Calcio/sangre , Enfermedades Cardiovasculares/patología , Humanos , Fallo Renal Crónico/patología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fosfatos/sangre
16.
Nephrol Dial Transplant ; 28(1): 62-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23045434

RESUMEN

BACKGROUND: Chronic kidney disease-mineral bone disorder (CKD-MBD) is a systemic syndrome characterized by imbalances in mineral homeostasis, renal osteodystrophy (ROD) and ectopic calcification. The mechanisms underlying this syndrome in individuals with chronic kidney disease (CKD) are not yet clear. METHODS: We examined the effect of normal phosphate (NP) or high phosphate (HP) feeding in the setting of CKD on bone pathology, serum biochemistry and vascular calcification in calcification-prone dilute brown non-agouti (DBA/2) mice. RESULTS: In both NP and HP-fed CKD mice, elevated serum parathyroid hormone and alkaline phosphatase (ALP) levels were observed, but serum phosphorus levels were equivalent compared with sham controls. CKD mice on NP diet showed trabecular alterations in the long bone consistent with high-turnover ROD, including increased trabecular number with abundant osteoblasts and osteoclasts. Despite trabecular bone and serum biochemical changes, CKD/NP mice did not develop vascular calcification. In contrast, CKD/HP mice developed arterial medial calcification (AMC), more severe trabecular bone alterations and cortical bone abnormalities that included decreased cortical thickness and density, and increased cortical porosity. Cortical bone porosity and trabecular number strongly correlated with the degree of aortic calcification. CONCLUSIONS: HP feeding was required to induce the full spectrum of CKD-MBD symptoms in CKD mice.


Asunto(s)
Densidad Ósea/fisiología , Enfermedades Óseas/fisiopatología , Huesos/patología , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/fisiopatología , Fosfatos/administración & dosificación , Insuficiencia Renal Crónica/fisiopatología , Calcificación Vascular/fisiopatología , Animales , Enfermedades Óseas/patología , Huesos/fisiopatología , Femenino , Ratones , Ratones Endogámicos DBA , Insuficiencia Renal Crónica/complicaciones
18.
Sci Rep ; 13(1): 9055, 2023 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-37270571

RESUMEN

Periodontal ligament stem cells (PDLSCs) play a significant role on periodontal tissue and alveolar bone homeostasis. During inflammation, interleukin (IL)-6 serves as one of key cytokine players controlling tissue reaction as well as alveolar bone tissue remodeling. It is believed that periodontal tissue inflammation causes periodontium degradation, especially alveolar bone. However, in this study, we show that an inflammatory mediator, IL-6, may serve another direction on alveolar bone homeostasis during inflammatory condition. We found that, IL-6 at 10 and 20 ng/mL was not cytotoxic and dose-dependently exerted beneficial effects on osteogenic differentiation of human PDLSCs (hPDLSCs), as demonstrated by increased alkaline phosphatase activity, mRNA expression of osteogenic markers, and matrix mineralization. The presence of physiological and inflammatory level of IL-6, the osteogenic differentiation potential by hPDLSCs was enhanced by several possible mechanisms including transforming growth factor (TGF), Wnt, and Notch pathways. After in-depth and thorough exploration, we found that Wnt pathway serves as key regulator controlling osteogenic differentiation by hPDLSCs amid the IL-6 presentation. Surprisingly, apart from other mesenchymal stem cells, distinct Wnt components are employed by hPDLSCs, and both canonical and non-canonical Wnt pathways are triggered by different mechanisms. Further validation by gene silencing, treatment with recombinant Wnt ligands, and ß-catenin stabilization/translocation confirmed that IL-6 governed the canonical Wnt/ß-catenin pathway via either WNT2B or WNT10B and employed WNT5A to activate the non-canonical Wnt pathway. These findings fulfill the homeostasis pathway governing periodontal tissue and alveolar bone regeneration and may serve for further therapeutic regimen design for restoring the tissues.


Asunto(s)
Osteogénesis , Ligamento Periodontal , Humanos , Interleucina-6/metabolismo , beta Catenina/metabolismo , Células Madre/metabolismo , Vía de Señalización Wnt/fisiología , Inflamación/metabolismo , Factores Inmunológicos/metabolismo , Diferenciación Celular , Células Cultivadas
19.
Res Sq ; 2023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-37461708

RESUMEN

Background: Ever since their discovery, induced pluripotent stem cells (iPSCs) have been extensively differentiated into a large variety of cell types. However, a limited amount of work has been dedicated to differentiating iPSCs into osteoclasts. While several differentiation protocols have been published, it remains unclear which protocols or differentiation methods are preferrable regarding the differentiation of osteoclasts. Methods: In this study we compare the osteoclastogenesis capacity of a peripheral blood mononuclear cell (PBMC)-derived iPSC line to a fibroblast-derived iPSC line in conjunction with either embryoid body-based or monolayer-based differentiation strategies. Both cell lines and differentiation protocols were investigated regarding their ability to generate osteoclasts and their inherent robustness and ease of use. The ability of both cell lines to remain undifferentiated while propagating using a feeder-free system was assessed using alkaline phosphatase staining. This was followed by evaluating mesodermal differentiation and the characterization of hematopoietic progenitor cells using flow cytometry. Finally, osteoclast yield and functionality based on resorptive activity, Cathepsin K and tartrate-resistant acid phosphatase (TRAP) expression were assessed. Results were validated using qRT-PCR throughout the differentiation stages. Results: Embryoid-body based differentiation yielded CD45+, CD14+, CD11b+ subpopulations which in turn differentiated into osteoclasts which demonstrated TRAP positivity, Cathepsin K expression and mineral resorptive capabilities. This was regardless of which iPSC line was used. Monolayer-based differentiation yielded lower quantities of hematopoietic cells that were mostly CD34+ and did not subsequently differentiate into osteoclasts. Conclusions: The outcome of this study demonstrates the successful differentiation of osteoclasts from iPSCs in conjunction with the embryoid-based differentiation method, while the monolayer-based method did not yield osteoclasts. No differences were observed regarding osteoclast differentiation between the PBMC and fibroblast-derived iPSC lines.

20.
Stem Cell Res Ther ; 14(1): 319, 2023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37936199

RESUMEN

BACKGROUND: Ever since their discovery, induced pluripotent stem cells (iPSCs) have been extensively differentiated into a large variety of cell types. However, a limited amount of work has been dedicated to differentiating iPSCs into osteoclasts. While several differentiation protocols have been published, it remains unclear which protocols or differentiation methods are preferable regarding the differentiation of osteoclasts. METHODS: In this study, we compared the osteoclastogenesis capacity of a peripheral blood mononuclear cell (PBMC)-derived iPSC line to a fibroblast-derived iPSC line in conjunction with either embryoid body-based or monolayer-based differentiation strategies. Both cell lines and differentiation protocols were investigated regarding their ability to generate osteoclasts and their inherent robustness and ease of use. The ability of both cell lines to remain undifferentiated while propagating using a feeder-free system was assessed using alkaline phosphatase staining. This was followed by evaluating mesodermal differentiation and the characterization of hematopoietic progenitor cells using flow cytometry. Finally, osteoclast yield and functionality based on resorptive activity, Cathepsin K and tartrate-resistant acid phosphatase (TRAP) expression were assessed. The results were validated using qRT-PCR throughout the differentiation stages. RESULTS: Embryoid body-based differentiation yielded CD45+, CD14+, CD11b+ subpopulations which in turn differentiated into osteoclasts which demonstrated TRAP positivity, Cathepsin K expression and mineral resorptive capabilities. This was regardless of which iPSC line was used. Monolayer-based differentiation yielded lower quantities of hematopoietic cells that were mostly CD34+ and did not subsequently differentiate into osteoclasts. CONCLUSIONS: The outcome of this study demonstrates the successful differentiation of osteoclasts from iPSCs in conjunction with the embryoid-based differentiation method, while the monolayer-based method did not yield osteoclasts. No differences were observed regarding osteoclast differentiation between the PBMC and fibroblast-derived iPSC lines.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Osteoclastos , Leucocitos Mononucleares , Catepsina K/metabolismo , Diferenciación Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA