Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Development ; 149(7)2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35297993

RESUMEN

Beige adipocytes have a discrete developmental origin and possess notable plasticity in their thermogenic capacity in response to various environmental cues, but the transcriptional machinery controlling beige adipocyte development and thermogenesis remains largely unknown. By analyzing beige adipocyte-specific knockout mice, we identified a transcription factor, forkhead box P4 (FOXP4), that differentially governs beige adipocyte differentiation and activation. Depletion of Foxp4 in progenitor cells impaired beige cell early differentiation. However, we observed that ablation of Foxp4 in differentiated adipocytes profoundly potentiated their thermogenesis capacity upon cold exposure. Of note, the outcome of Foxp4 deficiency on UCP1-mediated thermogenesis was confined to beige adipocytes, rather than to brown adipocytes. Taken together, we suggest that FOXP4 primes beige adipocyte early differentiation, but attenuates their activation by potent transcriptional repression of the thermogenic program.


Asunto(s)
Adipocitos Beige , Adipocitos Marrones , Animales , Diferenciación Celular/genética , Regulación de la Expresión Génica , Ratones , Termogénesis/genética
2.
Proc Natl Acad Sci U S A ; 115(35): 8799-8804, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-30104377

RESUMEN

Fundamental human traits, such as language and bipedalism, are associated with a range of anatomical adaptations in craniofacial shaping and skeletal remodeling. However, it is unclear how such morphological features arose during hominin evolution. FOXP2 is a brain-expressed transcription factor implicated in a rare disorder involving speech apraxia and language impairments. Analysis of its evolutionary history suggests that this gene may have contributed to the emergence of proficient spoken language. In the present study, through analyses of skeleton-specific knockout mice, we identified roles of Foxp2 in skull shaping and bone remodeling. Selective ablation of Foxp2 in cartilage disrupted pup vocalizations in a similar way to that of global Foxp2 mutants, which may be due to pleiotropic effects on craniofacial morphogenesis. Our findings also indicate that Foxp2 helps to regulate strength and length of hind limbs and maintenance of joint cartilage and intervertebral discs, which are all anatomical features that are susceptible to adaptations for bipedal locomotion. In light of the known roles of Foxp2 in brain circuits that are important for motor skills and spoken language, we suggest that this gene may have been well placed to contribute to coevolution of neural and anatomical adaptations related to speech and bipedal locomotion.


Asunto(s)
Remodelación Ósea/genética , Factores de Transcripción Forkhead , Locomoción/genética , Mutación , Proteínas Represoras , Cráneo/metabolismo , Vocalización Animal , Animales , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Miembro Posterior/metabolismo , Humanos , Ratones , Ratones Noqueados , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
4.
Proc Natl Acad Sci U S A ; 109(9): 3359-64, 2012 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-22331870

RESUMEN

The connexin 43 (Cx43) hemichannel (HC) in the mechanosensory osteocytes is a major portal for the release of factors responsible for the anabolic effects of mechanical loading on bone formation and remodeling. However, little is known about how the Cx43 molecule responds to mechanical stimulation leading to the opening of the HC. Here, we demonstrate that integrin α5ß1 interacts directly with Cx43 and that this interaction is required for mechanical stimulation-induced opening of the Cx43 HC. Direct mechanical perturbation via magnetic beads or conformational activation of integrin α5ß1 leads to the opening of the Cx43 HC, and this role of the integrin is independent of its association with an extracellular fibronectin substrate. PI3K signaling is responsible for the shear stress-induced conformational activation of integrin α5ß1 leading to the opening of the HC. These results identify an unconventional function of integrin that acts as a mechanical tether to induce opening of the HC and provide a mechanism connecting the effect of mechanical forces directly to anabolic function of the bone.


Asunto(s)
Conexina 43/metabolismo , Integrina alfa5beta1/fisiología , Osteocitos/metabolismo , Estrés Mecánico , Androstadienos/farmacología , Animales , Línea Celular , Cromonas/farmacología , Proteínas de la Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Separación Inmunomagnética , Integrina alfa5beta1/antagonistas & inhibidores , Ratones , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Mapeo de Interacción de Proteínas , ARN Interferente Pequeño/farmacología , Wortmanina
5.
J Cell Biochem ; 115(3): 450-6, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24142423

RESUMEN

The genome is under constant attack by self-produced reactive oxygen species and genotoxic reagents in the environment. Cells have evolved a DNA damage response (DDR) system to sense DNA damage, to halt cell cycle progression and repair the lesions, or to induce apoptosis if encountering irreparable damage. The best studied DDR pathways are the PIKK-p53 and PIKK-Chk1/2. Mutations in these genes encoding DDR molecules usually lead to genome instability and tumorigenesis. It is worth noting that there exist unconventional pathways that facilitate the canonical pathways or take over in the absence of the canonical pathways in DDR. This review will summarize on several unconventional pathways that participate in DDR with an emphasis on the BMP-Smad1 pathway, a known regulator of mouse development and bone remodeling.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Daño del ADN/genética , Neoplasias/genética , Proteína Smad1/genética , Animales , Apoptosis/genética , Proteínas Morfogenéticas Óseas/genética , Remodelación Ósea/genética , Reparación del ADN/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/genética , Proteína Smad1/biosíntesis , Proteína Smad1/metabolismo
6.
Stem Cell Res Ther ; 14(1): 188, 2023 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-37507770

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) are widely used in a variety of tissue regeneration and clinical trials due to their multiple differentiation potency. However, it remains challenging to maintain their replicative capability during in vitro passaging while preventing their premature cellular senescence. Forkhead Box P1 (FOXP1), a FOX family transcription factor, has been revealed to regulate MSC cell fate commitment and self-renewal capacity in our previous study. METHODS: Mass spectra analysis was performed to identify acetylation sites in FOXP1 protein. Single and double knockout mice of FOXP1 and HDAC7 were generated and analyzed with bone marrow MSCs properties. Gene engineering in human embryonic stem cell (hESC)-derived MSCs was obtained to evaluate the impact of FOXP1 key modification on MSC self-renewal potency. RESULTS: FOXP1 is deacetylated and potentiated by histone deacetylase 7 (HDAC7) in MSCs. FOXP1 and HDAC7 cooperatively sustain bone marrow MSC self-renewal potency while attenuating their cellular senescence. A mutation within human FOXP1 at acetylation site (T176G) homologous to murine FOXP1 T172G profoundly augmented MSC expansion capacity during early passages. CONCLUSION: These findings reveal a heretofore unanticipated mechanism by which deacetylation of FOXP1 potentiates self-renewal of MSC and protects them from cellular senescence. Acetylation of FOXP1 residue T172 as a critical modification underlying MSC proliferative capacity. We suggest that in vivo gene editing of FOXP1 may provide a novel avenue for manipulating MSC capability during large-scale expansion in clinical trials.


Asunto(s)
Senescencia Celular , Células Madre Mesenquimatosas , Animales , Humanos , Ratones , Diferenciación Celular/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Histona Desacetilasas/genética , Células Madre Mesenquimatosas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
7.
J Bone Miner Res ; 36(10): 2017-2026, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34131944

RESUMEN

Adiponectin (AdipoQ), a hormone abundantly secreted by adipose tissues, has multiple beneficial functions, including insulin sensitization as well as lipid and glucose metabolism. It has been reported that bone controls energy metabolism through an endocrine-based mechanism. In this study, we observed that bone also acts as an important endocrine source for AdipoQ, and its capacity in osteoblasts is controlled by the forkhead box P1 (FOXP1) transcriptional factor. Deletion of the Foxp1 gene in osteoblasts led to augmentation of AdipoQ levels accompanied by fueled energy expenditure in adipose tissues. In contrast, overexpression of Foxp1 in bones impaired AdipoQ secretion and restrained energy consumption. Chromatin immunoprecipitation sequencing (ChIP-seq) analysis revealed that AdipoQ expression, which increases as a function of bone age, is directly controlled by FOXP1. Our results indicate that bones, especially aged bones, provide an important source of a set of endocrine factors, including AdipoQ, that control body metabolism. © 2021 American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Tejido Adiposo , Metabolismo Energético , Tejido Adiposo/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Osteoblastos/metabolismo
8.
J Nutr ; 140(10): 1781-7, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20739449

RESUMEN

Bone is progressively lost with advancing age. Therapies are limited and the only effective proanabolic regimen presently available to restore bone is intermittent treatment with teriparatide (parathyroid hormone 1-34). Recent evidence suggests that dietary supplementation with dried plum (DP) can prevent bone loss due to estrogen deficiency. To determine whether dietary DP supplementation can prevent the loss of bone with aging and whether bone that has already been lost can be restored, adult (6 mo) and old (18 mo) male mice were fed a normal diet or isoenergetic, isonitrogenous diets supplemented with DP (0, 15, and 25% DP by weight) for 6 mo. MicroCT analysis and bone histomorphometry were used to assess bone volume, structure, and metabolic activity before, during, and after dietary supplementation. Mice fed the 0% DP diet (control diet) lost bone, whereas both adult and old mice fed the 25% DP-supplemented diet gained bone. Adult but not old mice fed the 15% diet also gained bone. Cancellous bone volume in mice receiving 25% DP exceeded baseline levels by 40-50%. Trabecular structure varied with diet and age and responses in old mice were generally blunted. Trabecular, but not cortical, mineral density varied with age and measures of bone anabolic activity were lower in aged mice. Our findings suggest that DP contains proanabolic factors that can dramatically increase bone volume and restore bone that has already been lost due to aging. In turn, DP may provide effective prophylactic and therapeutic agents for the treatment of osteoporosis.


Asunto(s)
Envejecimiento , Dieta , Alimentos en Conserva , Frutas , Osteoporosis/prevención & control , Prunus , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Densidad Ósea , Huesos/metabolismo , Huesos/patología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Fitoterapia
9.
Nat Commun ; 10(1): 5070, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31699980

RESUMEN

ß-Adrenergic receptor (ß-AR) signaling is a pathway controlling adaptive thermogenesis in brown or beige adipocytes. Here we investigate the biological roles of the transcription factor Foxp1 in brown/beige adipocyte differentiation and thermogenesis. Adipose-specific deletion of Foxp1 leads to an increase of brown adipose activity and browning program of white adipose tissues. The Foxp1-deficient mice show an augmented energy expenditure and are protected from diet-induced obesity and insulin resistance. Consistently, overexpression of Foxp1 in adipocytes impairs adaptive thermogenesis and promotes diet-induced obesity. A robust change in abundance of the ß3-adrenergic receptor (ß3-AR) is observed in brown/beige adipocytes from both lines of mice. Molecularly, Foxp1 directly represses ß3-AR transcription and regulates its desensitization behavior. Taken together, our findings reveal Foxp1 as a master transcriptional repressor of brown/beige adipocyte differentiation and thermogenesis, and provide an important clue for its targeting and treatment of obesity.


Asunto(s)
Adipocitos Beige/metabolismo , Adipocitos Marrones/metabolismo , Adipogénesis/genética , Metabolismo Energético/genética , Factores de Transcripción Forkhead/genética , Receptores Adrenérgicos beta 3/genética , Proteínas Represoras/genética , Termogénesis/genética , Tejido Adiposo Blanco/metabolismo , Animales , Dieta Alta en Grasa , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Humanos , Resistencia a la Insulina , Ratones , Obesidad/genética , Obesidad/metabolismo , Epiplón/metabolismo , Feocromocitoma/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Proteínas Represoras/metabolismo
10.
Biochim Biophys Acta ; 1770(8): 1169-80, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17570595

RESUMEN

Trichosanthin (TCS), a traditional Chinese medicine, exerts antitumor activities by inducing apoptosis in many different tumor cell lines. However, the mechanisms remain obscure. The present study focused on various caspase pathways that may be involved in TCS-induced apoptosis in leukemia HL-60 cells. Key caspases in both intrinsic and extrinsic pathways including caspase-8, -9 and -3 were activated upon TCS treatment. Additionally, TCS treatment induced upregulation of BiP and CHOP and also activated caspase-4, which for the first time strongly supported the involvement of endoplasmic reticulum stress pathway in TCS-induced apoptosis. Interestingly, although caspase-8 was activated, Fas/Fas ligand pathway was not involved as evidenced by a lack of induction of Fas or Fas ligand and a lack of inhibitory effect of anti-Fas blocking antibody on TCS-induced apoptosis. Instead, caspase-8 was activated in a caspase-9 and -4 dependent manner. The involvement of mitochondria was demonstrated by the reduction of mitochondrial membrane potential and release of cytochrome c and Smac besides the activation of caspase-9. Further investigation confirmed that caspase-3 was the major executioner caspase downstream to caspase-9, -4 and -8. Taken together, our results suggested that TCS-induced apoptosis in HL-60 cells was mainly mediated by mitochondrial and ER stress signaling pathways via caspase-3.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Transducción de Señal , Tricosantina/farmacología , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Caspasas Iniciadoras , Relación Dosis-Respuesta a Droga , Chaperón BiP del Retículo Endoplásmico , Activación Enzimática/efectos de los fármacos , Células HL-60 , Proteínas de Choque Térmico/metabolismo , Humanos , Potenciales de la Membrana/efectos de los fármacos , Modelos Biológicos , Chaperonas Moleculares/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factor de Transcripción CHOP/metabolismo
11.
Biochim Biophys Acta ; 1770(1): 63-70, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16949756

RESUMEN

Trichosanthin (TCS), a type I ribosome-inactivating protein, induces cell death in various cell types including several tumor cell lines. However, the mechanism remains largely uncharacterized. In this study, we investigated the possible mechanism underlying its cytotoxicity by using human chronic myeloid leukemia cell line K562. We found that TCS induced apoptosis in K562 cells in a time- and concentration-dependent manner and can be blocked by caspase-3 inhibitors. Interestingly, TCS treatment induced a transient elevation in intracellular calcium concentration and a slow increase in reactive oxygen species production, while calcium chelators and antioxidants had no obvious effect on TCS-induced apoptosis, suggesting that calcium changes and reactive oxygen species may not be involved in TCS-mediated apoptosis in K562 cells. Instead we found that TCS partly inhibited PKC activity. Indeed, the PKC activator, PMA, inhibited while the PKC inhibitor, calphostin c, enhanced TCS-induced apoptosis. These PKC modulators had similar effects on TCS-induced cleavage of caspase-3, and caspase-3 inhibitors prevented calphostin c-enhanced apoptosis induced by TCS. In summary, we conclude that TCS induces apoptosis in K562 cells partly via PKC inhibition and caspase-3 activation.


Asunto(s)
Apoptosis/efectos de los fármacos , Naftalenos/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Tricosantina/farmacología , Calcio/metabolismo , Señalización del Calcio , Caspasa 3/metabolismo , Citometría de Flujo , Humanos , Microscopía Confocal , Especies Reactivas de Oxígeno
12.
Endocrinology ; 159(1): 260-271, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29069393

RESUMEN

Sclerostin antibody (Scl-Ab) restored bone mass and strength in the ovariectomized rat model of postmenopausal osteoporosis. Increased bone mineral density (BMD) and decreased skeletal fragility fracture risk have been reported in postmenopausal osteoporotic women receiving Scl-Ab. In males, loss of androgen leads to rapid decreases in BMD and an increased risk of fragility fractures. We hypothesized that Scl-Ab could reverse the loss of bone mass and strength caused by androgen ablation in the orchiectomized (ORX) rat model of male osteoporosis. We treated 9-month-old ORX Sprague Dawley rats (3 months after ORX) subcutaneously twice weekly with vehicle or Scl-Ab (5 or 25 mg/kg) for 6 weeks (n = 10 per group). Both doses of Scl-Ab fully reversed the BMD deficit in the lumbar spine and femur and tibia in ORX rats. Microcomputed tomography showed that the bone mass in the fifth lumbar vertebral body, femur diaphysis, and femoral neck were dose-dependently restored by Scl-Ab. The bone strength at these sites increased significantly with Scl-Ab to levels matching those of sham-operated controls and correlated positively with improvements in bone mineral content, demonstrating bone quality maintenance. Dynamic histomorphometry of the tibial diaphysis and second lumbar vertebral body demonstrated that Scl-Ab significantly increased bone formation on periosteal, endocortical, and trabecular surfaces and significantly decreased bone resorption on endocortical and trabecular surfaces. The effects of Scl-Ab on increasing bone formation and decreasing bone resorption led to restoration of bone mass and strength in androgen-deficient rats. These findings support the ongoing evaluation of Scl-Ab as a potential therapeutic agent for osteoporosis in men.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Conservadores de la Densidad Ósea/uso terapéutico , Proteínas Morfogenéticas Óseas/inmunología , Huesos/efectos de los fármacos , Modelos Animales de Enfermedad , Marcadores Genéticos/inmunología , Osteogénesis/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Absorciometría de Fotón , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Densidad Ósea/efectos de los fármacos , Conservadores de la Densidad Ósea/administración & dosificación , Conservadores de la Densidad Ósea/efectos adversos , Proteínas Morfogenéticas Óseas/metabolismo , Resorción Ósea/prevención & control , Huesos/diagnóstico por imagen , Huesos/patología , Relación Dosis-Respuesta a Droga , Inyecciones Subcutáneas , Masculino , Terapia Molecular Dirigida , Orquiectomía , Tamaño de los Órganos/efectos de los fármacos , Osteoporosis/diagnóstico por imagen , Osteoporosis/patología , Próstata/efectos de los fármacos , Próstata/patología , Ratas Sprague-Dawley , Vesículas Seminales/efectos de los fármacos , Vesículas Seminales/patología , Resistencia al Corte/efectos de los fármacos , Aumento de Peso/efectos de los fármacos , Microtomografía por Rayos X
13.
Sleep ; 40(12)2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29029290

RESUMEN

Study Objectives: The correlated activation of pre- and postsynaptic neurons is essential for the NMDA receptor-mediated Ca2+ influx by removing Mg2+ from block site and NMDA receptors have been implicated in phase resetting of circadian clocks. So we assessed rest:activity rhythms in Mg2+ block defective animals. Methods: Using Drosophila locomotor monitoring system, we checked circadian rest:activity rhythms of different mutants under constant darkness (DD) and light:dark (LD) conditions. We recorded NMDA receptor-mediated currents or Ca2+ increase in neurons using patch-clamp and Ca2+ imaging techniques. Results: We found that Mg2+ block defective mutant flies were completely arrhythmic under DD. To further understand the role of Mg2+ block in daily circadian rest:activity, we observed the mutant files under LD cycles, and we found severely reduced morning anticipation and advanced evening peak compared to control flies. We also used tissue-specific expression of Mg2+ block defective NMDA receptors and demonstrated pigment-dispersing factor receptor (PDFR)-expressing circadian neurons were implicated in mediating the circadian rest:activity deficits. Endogenous functional NMDA receptors are expressed in most Drosophila neurons, including in a subgroup of dorsal neurons (DN1s). Subsequently, we determined that the uncorrelated extra Ca2+ influx may act in part through Ca2+/Calmodulin (CaM)-stimulated PDE1c pathway leading to morning behavior phenotypes. Conclusions: These results demonstrate that Mg2+ block of NMDA receptors at resting potential is essential for the daily circadian rest:activity rhythms and we propose that Mg2+ block functions to suppress CaM-stimulated PDE1c activation at resting potential, thus regulating Ca2+ and cyclic AMP oscillations in circadian and sleep circuits.


Asunto(s)
Señalización del Calcio/fisiología , Ritmo Circadiano/fisiología , Proteínas de Drosophila/fisiología , Magnesio/fisiología , Actividad Motora/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Animales , Animales Modificados Genéticamente , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Relojes Circadianos/fisiología , Ritmo Circadiano/efectos de los fármacos , Oscuridad , Drosophila , Magnesio/farmacología , Actividad Motora/efectos de los fármacos , Mutación/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fotoperiodo , Descanso/fisiología
14.
J Bone Miner Res ; 32(7): 1406-1420, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28370465

RESUMEN

Vitamin D is involved in a range of physiological processes and its active form and analogs have been used to treat diseases such as osteoporosis. Yet how vitamin D executes its function remains unsolved. Here we show that the active form of vitamin D calcitriol increases the peak bone mass in mice by inhibiting osteoclastogenesis and bone resorption. Although calcitriol modestly promoted osteoclast maturation, it strongly inhibited osteoclast lineage commitment from its progenitor monocyte by increasing Smad1 transcription via the vitamin D receptor and enhancing BMP-Smad1 activation, which in turn led to increased IκBα expression and decreased NF-κB activation and NFATc1 expression, with IκBα being a Smad1 target gene. Inhibition of BMP type I receptor or ablation of Bmpr1a in monocytes alleviated the inhibitory effects of calcitriol on osteoclast commitment, bone resorption, and bone mass augmentation. These findings uncover crosstalk between the BMP-Smad1 and RANKL-NF-κB pathways during osteoclastogenesis that underlies the action of active vitamin D on bone health. © 2017 American Society for Bone and Mineral Research.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Resorción Ósea/tratamiento farmacológico , Calcitriol/farmacología , Proteínas I-kappa B/metabolismo , FN-kappa B/metabolismo , Osteoclastos/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína Smad1/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/genética , Resorción Ósea/genética , Resorción Ósea/metabolismo , Proteínas I-kappa B/genética , Ratones , Ratones Transgénicos , FN-kappa B/genética , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Transducción de Señal/genética , Proteína Smad1/genética
15.
Stem Cell Reports ; 8(6): 1600-1616, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28479301

RESUMEN

The bone size and quality, acquired during adolescent growth under the influence of anabolic hormones, growth factors, and nutrients, determine the height and bone stability and forecast osteoporosis risks in late life. Yet bone size and quality control mechanisms remain enigmatic. To study the roles of mammalian target of rapamycin (mTOR) signaling, sensor of growth factors and nutrients, in bone size and quality regulation, we ablated Tsc1, a suppressor of mTOR, in mesenchymal stromal cells (MSCs), monocytes, or their progenies osteoblasts and osteoclasts. mTOR activation in MSCs, but much less in osteoblasts, increased bone width and mass due to MSC hyperproliferation, but decreased bone length and mineral contents due to defective MSC differentiation. mTOR activation promotes bone mineral accretion by inhibiting osteoclast differentiation and activity directly or via coupling with MSCs. Tuberous sclerosis complex patient studies confirmed these findings. Thus, mTOR regulates bone size via MSCs and bone quality by suppressing catabolic activities of osteoclasts.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Osteoclastos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Esclerosis Tuberosa/diagnóstico , Adolescente , Adulto , Animales , Densidad Ósea , Huesos/diagnóstico por imagen , Calcificación Fisiológica , Diferenciación Celular , Células Cultivadas , Niño , Preescolar , Condrogénesis , Colágeno Tipo I/sangre , Femenino , Fémur/metabolismo , Proteínas de Homeodominio/genética , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Noqueados , Persona de Mediana Edad , Monocitos/citología , Monocitos/metabolismo , Osteocalcina/sangre , Osteoclastos/citología , Osteogénesis , Péptidos/sangre , Esclerosis Tuberosa/metabolismo , Esclerosis Tuberosa/patología , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Microtomografía por Rayos X , Adulto Joven
16.
J Biochem ; 139(4): 725-31, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16672273

RESUMEN

Trichosanthin is the active protein component in the Chinese herb Trichosanthes kirilowi, which has distinct pharmacological properties. The cytotoxicity of trichosanthin was demonstrated by its selective inhibition of various choriocarcinoma cells. When Jar cells were treated with trichosanthin, the influx of calcium into the cells was observed by confocal laser scanning microscopy. When the distribution of trichosanthin-binding proteins on Jar cells was studied, two classes of binding sites for trichosanthin were shown by radioligand binding assay. Furthermore, the cytoplasmic membrane of Jar cells was biotinylated and the trichosanthin-binding proteins were isolated with trichosanthin-coupled Sepharose beads. Two protein bands with molecular masses of about 50 kDa and 60 kDa were revealed, further characterization of which should shed light on the mechanism of the selective cytotoxicity of trichosanthin to Jar cells.


Asunto(s)
Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Tricosantina/metabolismo , Animales , Antineoplásicos Fitogénicos/metabolismo , Antineoplásicos Fitogénicos/toxicidad , Biotinilación , Calcio/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/genética , Línea Celular , Línea Celular Tumoral , Membrana Celular/química , Supervivencia Celular/efectos de los fármacos , Coriocarcinoma/metabolismo , Coriocarcinoma/patología , Coriocarcinoma/ultraestructura , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Immunoblotting , Microscopía Confocal , Proteínas de Plantas/metabolismo , Proteínas de Plantas/toxicidad , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tricosantina/toxicidad , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patología , Neoplasias Uterinas/ultraestructura
17.
Exp Biol Med (Maywood) ; 240(8): 1099-106, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26088863

RESUMEN

The aging population and the incidence of aging-related diseases such as osteoporosis are on the rise. Aging at the tissue and organ levels usually involves tissue stem cells. Human and animal model studies indicate that aging affects two aspects of mesenchymal stem cell (MSC): a decrease in the bone marrow MSC pool and biased differentiation into adipocyte at the cost of osteoblast, which underlie the etiology of osteoporosis. Aging of MSC cells is also detrimental to some non-skeletal tissues, in particular the hematopoietic system, where MSCs serve as a niche component. In addition, aging compromises the therapeutic potentials of MSC cells, including cells isolated from aged individuals or cells cultured for many passages. Here we discuss the recent progress on our understanding of MSC aging, with a focus on the effects of MSC aging on bone remodeling and hematopoiesis and the mechanisms of MSC aging.


Asunto(s)
Médula Ósea/metabolismo , Remodelación Ósea/fisiología , Senescencia Celular/fisiología , Hematopoyesis/fisiología , Células Madre Mesenquimatosas/metabolismo , Nicho de Células Madre/fisiología , Animales , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología
18.
J Mol Cell Biol ; 7(2): 105-18, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25757624

RESUMEN

The DNA damage response helps to maintain genome integrity, suppress tumorigenesis, and mediate the effects of radiotherapy and chemotherapy. Our previous studies have shown that Smad1 is upregulated and activated by Atm in DNA damage response, which can further bind to p53 and promote p53 stabilization. Here we report another aspect of the interplay between p53 and Smad1. Comparison of rectal tumor against paired paraneoplastic specimens and analysis of >500 colorectal tumors revealed that Smad1 was upregulated in tumor samples, which was attributable to p53 defects. Using MEFs as a model, we found that knockdown of the elevated Smad1 in p53(-/-) MEFs promoted cell proliferation, E1A/Ras-induced cell transformation, and tumorigenesis. Mechanistic studies suggest that elevated Smad1 and momentary activation inhibit cell proliferation by upregulating p57Kip2 and enhancing Atm-Chk2 activation. Surprisingly, elevated Smad1 appears to have a negative effect on chemotherapy, as colorectal tumors, primary cancer cells, and cell lines with Smad1 knockdown all showed an increase in chemosensitivity, which could be attributable to elevated p57Kip2. These findings underscore the significance of Smad1-p53 interaction in tumor suppression and reveal an unexpected role for Smad1 in chemoresistance of colorectal cancers.


Asunto(s)
Carcinogénesis/metabolismo , Neoplasias del Recto/metabolismo , Proteína Smad1/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Secuencia de Aminoácidos , Animales , Antibióticos Antineoplásicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Carcinogénesis/genética , Proliferación Celular , Transformación Celular Neoplásica , Quinasa de Punto de Control 2/metabolismo , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Expresión Génica , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Datos de Secuencia Molecular , Trasplante de Neoplasias , Neoplasias del Recto/tratamiento farmacológico , Neoplasias del Recto/patología , Proteína Smad1/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba
19.
PLoS One ; 9(12): e113785, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25470749

RESUMEN

Platelet-derived growth factors (PDGFs) play important roles in skeletal development and bone fracture healing, yet how PDGFs execute their functions remains incompletely understood. Here we show that PDGF-AA, but not -AB or -BB, could activate the BMP-Smad1/5/8 pathway in mesenchymal stem cells (MSCs), which requires BMPRIA as well as PDGFRα. PDGF-AA promotes MSC osteogenic differentiation through the BMP-Smad1/5/8-Runx2/Osx axis and MSC migration via the BMP-Smad1/5/8-Twist1/Atf4 axis. Mechanistic studies show that PDGF-AA activates BMP-Smad1/5/8 signaling by feedback down-regulating PDGFRα, which frees BMPRI and allows for BMPRI-BMPRII complex formation to activate smad1/5/8, using BMP molecules in the microenvironment. This study unravels a physical and functional interaction between PDGFRα and BMPRI, which plays an important role in MSC differentiation and migration, and establishes a link between PDGF-AA and BMPs pathways, two essential regulators of embryonic development and tissue homeostasis.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Smad/metabolismo , Animales , Western Blotting , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/deficiencia , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Osteogénesis/genética , Unión Proteica , Interferencia de ARN , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal/efectos de los fármacos , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo
20.
Mol Cell Biol ; 30(1): 206-19, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19841066

RESUMEN

Gap junction intercellular communication in osteocytes plays an important role in bone remodeling in response to mechanical loading; however, the responsible molecular mechanisms remain largely unknown. Here, we show that phosphoinositide-3 kinase (PI3K)/Akt signaling activated by fluid flow shear stress and prostaglandin E(2) (PGE(2)) had a stimulatory effect on both connexin 43 (Cx43) mRNA and protein expression. PGE(2) inactivated glycogen synthase kinase 3 (GSK-3) and promoted nuclear localization and accumulation of beta-catenin. Knockdown of beta-catenin expression resulted in a reduction in Cx43 protein. Furthermore, the chromatin immunoprecipitation (ChIP) assay demonstrated an association of beta-catenin with the Cx43 promoter, suggesting that beta-catenin could regulate Cx43 expression at the level of gene transcription. We have previously reported that PGE(2) activates cyclic AMP (cAMP)-protein kinase A (PKA) signaling and increases Cx43 and gap junctions. Interestingly, the activation of PI3K/Akt appeared to be independent of the activation of PKA, whereas both PI3K/Akt and PKA signaling inactivated GSK-3 and increased beta-catenin translocation. Together, these results suggest that shear stress, through PGE(2) release, activates both PI3K/Akt and cAMP-PKA signaling, which converge through the inactivation of GSK-3, leading to the increase in nuclear accumulation of beta-catenin. beta-Catenin binds to the Cx43 promoter, stimulating Cx43 expression and functional gap junctions between osteocytes.


Asunto(s)
Conexina 43/metabolismo , Dinoprostona/fisiología , Uniones Comunicantes/fisiología , Glucógeno Sintasa Quinasa 3/fisiología , Osteocitos/fisiología , Transcripción Genética , beta Catenina/fisiología , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Núcleo Celular/metabolismo , Conexina 43/genética , AMP Cíclico/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Ratones , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal , beta Catenina/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA