Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hum Mol Genet ; 33(1): 33-37, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37738569

RESUMEN

Inhaled nitric oxide (NO) therapy has been reported to improve lung growth in premature newborns. However, the underlying mechanisms by which NO regulates lung development remain largely unclear. NO is enzymatically produced by three isoforms of nitric oxide synthase (NOS) enzymes. NOS knockout mice are useful tools to investigate NO function in the lung. Each single NOS knockout mouse does not show obvious lung alveolar phenotype, likely due to compensatory mechanisms. While mice lacking all three NOS isoforms display impaired lung alveolarization, implicating NO plays a pivotal role in lung alveolarization. Argininosuccinate lyase (ASL) is the only mammalian enzyme capable of synthesizing L-arginine, the sole precursor for NOS-dependent NO synthesis. ASL is also required for channeling extracellular L-arginine into a NO-synthetic complex. Thus, ASL deficiency (ASLD) is a non-redundant model for cell-autonomous, NOS-dependent NO deficiency. Here, we assessed lung alveolarization in ASL-deficient mice. Hypomorphic deletion of Asl (AslNeo/Neo) results in decreased lung alveolarization, accompanied with reduced level of S-nitrosylation in the lung. Genetic ablation of one copy of Caveolin-1, which is a negative regulator of NO production, restores total S-nitrosylation as well as lung alveolarization in AslNeo/Neo mice. Importantly, NO supplementation could partially rescue lung alveolarization in AslNeo/Neo mice. Furthermore, endothelial-specific knockout mice (VE-Cadherin Cre; Aslflox/flox) exhibit impaired lung alveolarization at 12 weeks old, supporting an essential role of endothelial-derived NO in the enhancement of lung alveolarization. Thus, we propose that ASLD is a model to study NO-mediated lung alveolarization.


Asunto(s)
Argininosuccinatoliasa , Óxido Nítrico , Animales , Ratones , Argininosuccinatoliasa/genética , Óxido Nítrico Sintasa/genética , Arginina/genética , Ratones Noqueados , Pulmón , Isoformas de Proteínas , Mamíferos
2.
Cell Chem Biol ; 30(9): 1002-1003, 2023 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-37738949

RESUMEN

Individuals with type 1 diabetes (T1D) have an increased risk of osteoporosis and fracture. In this issue of Cell Chemical Biology, Ji et al.1 show that impaired glucose metabolism in the bone-forming osteoblast drives diabetic osteoporosis in Akita mice, a mouse model of T1D.


Asunto(s)
Diabetes Mellitus Tipo 1 , Osteoporosis , Humanos , Animales , Ratones , Diabetes Mellitus Tipo 1/complicaciones , Glucólisis , Osteoblastos , Modelos Animales de Enfermedad
3.
JCI Insight ; 8(17)2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37490345

RESUMEN

Nitric oxide (NO) is a critical signaling molecule that has been implicated in the pathogenesis of neurocognitive diseases. Both excessive and insufficient NO production have been linked to pathology. Previously, we have shown that argininosuccinate lyase deficiency (ASLD) is a novel model system to investigate cell-autonomous, nitric oxide synthase-dependent NO deficiency. Humans with ASLD are at increased risk for developing hyperammonemia due to a block in ureagenesis. However, natural history studies have shown that individuals with ASLD have multisystem disease including neurocognitive deficits that can be independent of ammonia. Here, using ASLD as a model of NO deficiency, we investigated the effects of NO on brain endothelial cells in vitro and the blood-brain barrier (BBB) in vivo. Knockdown of ASL in human brain microvascular endothelial cells (HBMECs) led to decreased transendothelial electrical resistance, indicative of increased cell permeability. Mechanistically, treatment with an NO donor or inhibition of Claudin-1 improved barrier integrity in ASL-deficient HBMECs. Furthermore, in vivo assessment of a hypomorphic mouse model of ASLD showed increased BBB leakage, which was partially rescued by NO supplementation. Our results suggest that ASL-mediated NO synthesis is required for proper maintenance of brain microvascular endothelial cell functions as well as BBB integrity.


Asunto(s)
Aciduria Argininosuccínica , Ratones , Animales , Humanos , Aciduria Argininosuccínica/genética , Aciduria Argininosuccínica/metabolismo , Aciduria Argininosuccínica/patología , Óxido Nítrico/metabolismo , Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Claudinas/metabolismo , Modelos Animales de Enfermedad
4.
Dis Model Mech ; 16(8)2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37486182

RESUMEN

SLC7A7 deficiency, or lysinuric protein intolerance (LPI), causes loss of function of the y+LAT1 transporter critical for efflux of arginine, lysine and ornithine in certain cells. LPI is characterized by urea cycle dysfunction, renal disease, immune dysregulation, growth failure, delayed bone age and osteoporosis. We previously reported that Slc7a7 knockout mice (C57BL/6×129/SvEv F2) recapitulate LPI phenotypes, including growth failure. Our main objective in this study was to characterize the skeletal phenotype in these mice. Compared to wild-type littermates, juvenile Slc7a7 knockout mice demonstrated 70% lower body weights, 87% lower plasma IGF-1 concentrations and delayed skeletal development. Because poor survival prevents evaluation of mature knockout mice, we generated a conditional Slc7a7 deletion in mature osteoblasts or mesenchymal cells of the osteo-chondroprogenitor lineage, but no differences in bone architecture were observed. Overall, global Slc7a7 deficiency caused growth failure with low plasma IGF-1 concentrations and delayed skeletal development, but Slc7a7 deficiency in the osteoblastic lineage was not a major contributor to these phenotypes. Future studies utilizing additional tissue-specific Slc7a7 knockout models may help dissect cell-autonomous and non-cell-autonomous mechanisms underlying phenotypes in LPI.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina , Animales , Ratones , Sistema de Transporte de Aminoácidos y+L , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Noqueados
5.
Am J Hum Genet ; 108(9): 1710-1724, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34450031

RESUMEN

Coatomer complexes function in the sorting and trafficking of proteins between subcellular organelles. Pathogenic variants in coatomer subunits or associated factors have been reported in multi-systemic disorders, i.e., coatopathies, that can affect the skeletal and central nervous systems. We have identified loss-of-function variants in COPB2, a component of the coatomer complex I (COPI), in individuals presenting with osteoporosis, fractures, and developmental delay of variable severity. Electron microscopy of COPB2-deficient subjects' fibroblasts showed dilated endoplasmic reticulum (ER) with granular material, prominent rough ER, and vacuoles, consistent with an intracellular trafficking defect. We studied the effect of COPB2 deficiency on collagen trafficking because of the critical role of collagen secretion in bone biology. COPB2 siRNA-treated fibroblasts showed delayed collagen secretion with retention of type I collagen in the ER and Golgi and altered distribution of Golgi markers. copb2-null zebrafish embryos showed retention of type II collagen, disorganization of the ER and Golgi, and early larval lethality. Copb2+/- mice exhibited low bone mass, and consistent with the findings in human cells and zebrafish, studies in Copb2+/- mouse fibroblasts suggest ER stress and a Golgi defect. Interestingly, ascorbic acid treatment partially rescued the zebrafish developmental phenotype and the cellular phenotype in Copb2+/- mouse fibroblasts. This work identifies a form of coatopathy due to COPB2 haploinsufficiency, explores a potential therapeutic approach for this disorder, and highlights the role of the COPI complex as a regulator of skeletal homeostasis.


Asunto(s)
Huesos/metabolismo , Proteína Coat de Complejo I/genética , Proteína Coatómero/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Osteoporosis/genética , Animales , Ácido Ascórbico/farmacología , Huesos/efectos de los fármacos , Huesos/patología , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Niño , Preescolar , Proteína Coat de Complejo I/deficiencia , Proteína Coatómero/química , Proteína Coatómero/deficiencia , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Discapacidades del Desarrollo/diagnóstico por imagen , Discapacidades del Desarrollo/metabolismo , Discapacidades del Desarrollo/patología , Embrión no Mamífero , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación del Desarrollo de la Expresión Génica , Aparato de Golgi , Haploinsuficiencia , Humanos , Discapacidad Intelectual/diagnóstico por imagen , Discapacidad Intelectual/metabolismo , Discapacidad Intelectual/patología , Masculino , Ratones , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Osteoporosis/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Índice de Severidad de la Enfermedad , Pez Cebra
6.
J Clin Invest ; 131(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33373331

RESUMEN

Previous studies have shown that nitric oxide (NO) supplements may prevent bone loss and fractures in preclinical models of estrogen deficiency. However, the mechanisms by which NO modulates bone anabolism remain largely unclear. Argininosuccinate lyase (ASL) is the only mammalian enzyme capable of synthesizing arginine, the sole precursor for nitric oxide synthase-dependent (NOS-dependent) NO synthesis. Moreover, ASL is also required for channeling extracellular arginine to NOS for NO production. ASL deficiency (ASLD) is thus a model to study cell-autonomous, NOS-dependent NO deficiency. Here, we report that loss of ASL led to decreased NO production and impairment of osteoblast differentiation. Mechanistically, the bone phenotype was at least in part driven by the loss of NO-mediated activation of the glycolysis pathway in osteoblasts that led to decreased osteoblast differentiation and function. Heterozygous deletion of caveolin 1, a negative regulator of NO synthesis, restored NO production, osteoblast differentiation, glycolysis, and bone mass in a hypomorphic mouse model of ASLD. The translational significance of these preclinical studies was further reiterated by studies conducted in induced pluripotent stem cells from an individual with ASLD. Taken together, our findings suggest that ASLD is a unique genetic model for studying NO-dependent osteoblast function and that the NO/glycolysis pathway may be a new target to modulate bone anabolism.


Asunto(s)
Aciduria Argininosuccínica/metabolismo , Huesos/metabolismo , Diferenciación Celular , Glucólisis , Ácido Nítrico/metabolismo , Osteoblastos/metabolismo , Adolescente , Adulto , Animales , Aciduria Argininosuccínica/genética , Aciduria Argininosuccínica/patología , Huesos/patología , Niño , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Osteoblastos/patología
7.
Nature ; 582(7810): 134, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32483375

RESUMEN

A Retraction to this paper has been published and can be accessed via a link at the top of the paper.

8.
JCI Insight ; 5(4)2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-31990680

RESUMEN

BACKGROUNDLiver disease in urea cycle disorders (UCDs) ranges from hepatomegaly and chronic hepatocellular injury to cirrhosis and end-stage liver disease. However, the prevalence and underlying mechanisms are unclear.METHODSWe estimated the prevalence of chronic hepatocellular injury in UCDs using data from a multicenter, longitudinal, natural history study. We also used ultrasound with shear wave elastography and FibroTest to evaluate liver stiffness and markers of fibrosis in individuals with argininosuccinate lyase deficiency (ASLD), a disorder with high prevalence of elevated serum alanine aminotransferase (ALT). To understand the human observations, we evaluated the hepatic phenotype of the AslNeo/Neo mouse model of ASLD.RESULTSWe demonstrate a high prevalence of elevated ALT in ASLD (37%). Hyperammonemia and use of nitrogen-scavenging agents, 2 markers of disease severity, were significantly (P < 0.001 and P = 0.001, respectively) associated with elevated ALT in ASLD. In addition, ultrasound with shear wave elastography and FibroTest revealed increased echogenicity and liver stiffness, even in individuals with ASLD and normal aminotransferases. The AslNeo/Neo mice mimic the human disorder with hepatomegaly, elevated aminotransferases, and excessive hepatic glycogen noted before death (3-5 weeks of age). This excessive hepatic glycogen is associated with impaired hepatic glycogenolysis and decreased glycogen phosphorylase and is rescued with helper-dependent adenovirus expressing Asl using a liver-specific (ApoE) promoter.CONCLUSIONOur results link urea cycle dysfunction and impaired hepatic glucose metabolism and identify a mouse model of liver disease in the setting of urea cycle dysfunction.TRIAL REGISTRATIONThis study has been registered at ClinicalTrials.gov (NCT03721367, NCT00237315).FUNDINGFunding was provided by NIH, Burroughs Wellcome Fund, NUCDF, Genzyme/ACMG Foundation, and CPRIT.


Asunto(s)
Argininosuccinatoliasa/metabolismo , Hepatopatías/metabolismo , Glucógeno Hepático/metabolismo , Alanina Transaminasa/sangre , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Humanos , Hepatopatías/complicaciones , Hepatopatías/enzimología , Estudios Longitudinales , Ratones , Trastornos Innatos del Ciclo de la Urea/complicaciones
9.
Nature ; 570(7761): E51, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31127195

RESUMEN

Change history: In this Letter, the citation to 'Fig. 4e, f' in the main text should be 'Fig. 3e, f'. This has not been corrected online.

10.
JBMR Plus ; 2(4): 235-239, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30283904

RESUMEN

The heritable disorder osteogenesis imperfecta (OI) is characterized by bone fragility and low bone mass. OI type VI is an autosomal recessive form of the disorder with moderate to severe bone fragility. OI type VI is caused by mutations in the serpin peptidase inhibitor, clade F, member 1 (SERPINF1), the gene coding for pigment epithelium-derived factor (PEDF). Here, we report a patient with OI type VI caused by a novel homozygous intronic variant in SERPINF1 identified by whole-exome sequencing (WES). The mutation was not identified using a low bone mass gene panel based on next-generation sequencing. This variant creates a novel consensus splice donor site (AGGC to AGGT) in intron 4. Analysis of cDNA generated from fibroblasts revealed retention of a 32-bp intronic fragment between exons 4 and 5 in the cDNA, a result of alternative splicing from the novel splice-donor site. As a result, the aberrant insertion of this intronic fragment generated a frameshift pathogenic variant and induced nonsense-mediated decay. Furthermore, gene expression by quantitative PCR showed SERPINF1 expression was dramatically reduced in patient fibroblasts, and PEDF level was also significantly reduced in the patient's plasma. In conclusion, we report a novel homozygous variant that generates an alternative splice-donor in intron 4 of SERPINF1 which gives rise to severe bone fragility. The work also demonstrates clinical utility of WES analysis, and consideration of noncoding variants, in the diagnostic setting of rare bone diseases. © 2018 The Authors. JBMR Plus is published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.

11.
Am J Hum Genet ; 103(2): 276-287, 2018 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-30075114

RESUMEN

Primary hypertension is a major risk factor for ischemic heart disease, stroke, and chronic kidney disease. Insights obtained from the study of rare Mendelian forms of hypertension have been invaluable in elucidating the mechanisms causing primary hypertension and development of antihypertensive therapies. Endothelial cells play a key role in the regulation of blood pressure; however, a Mendelian form of hypertension that is primarily due to endothelial dysfunction has not yet been described. Here, we show that the urea cycle disorder, argininosuccinate lyase deficiency (ASLD), can manifest as a Mendelian form of endothelial-dependent hypertension. Using data from a human clinical study, a mouse model with endothelial-specific deletion of argininosuccinate lyase (Asl), and in vitro studies in human aortic endothelial cells and induced pluripotent stem cell-derived endothelial cells from individuals with ASLD, we show that loss of ASL in endothelial cells leads to endothelial-dependent vascular dysfunction with reduced nitric oxide (NO) production, increased oxidative stress, and impaired angiogenesis. Our findings show that ASLD is a unique model for studying NO-dependent endothelial dysfunction in human hypertension.


Asunto(s)
Argininosuccinatoliasa/genética , Aciduria Argininosuccínica/genética , Células Endoteliales/patología , Hipertensión/genética , Adolescente , Animales , Presión Sanguínea/genética , Células Cultivadas , Niño , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Neovascularización Patológica/genética , Óxido Nítrico/genética , Estrés Oxidativo/genética , Trastornos Innatos del Ciclo de la Urea/genética
12.
Mol Genet Metab ; 125(1-2): 112-117, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30055993

RESUMEN

Arginine is a semi-essential amino acid which serves as a substrate for nitric oxide (NO) production by nitric oxide synthase (NOS) and a precursor for various metabolites including ornithine, creatine, polyamines, and agmatine. Arginase competes with nitric oxide synthase for substrate arginine to produce orthinine and urea. There is contradictory evidence in the literature on the role of nitric oxide in the pathophysiology of traumatic brain injury (TBI). These contradictory perspectives are likely due to different NOS isoforms - endothelial (eNOS), inducible (iNOS) and neuronal (nNOS) which are expressed in the central nervous system. Of these, the role of nNOS in acute injury remains less clear. This study aimed to employ a genetic approach by overexpressing arginase isoforms specifically in neurons using a Thy-1 promoter to manipulate cell autonomous NO production in the context of TBI. The hypothesis was that increased arginase would divert arginine from pathological NO production. We generated 2 mouse lines that overexpress arginase I (a cytoplasmic enzyme) or arginase II (a mitochondrial enzyme) in neurons of FVB mice. We found that two-weeks after induction of controlled cortical injury, overexpressing arginase I but not arginase II in neurons significantly reduced contusion size and contusion index compared to wild-type (WT) mice. This study establishes enhanced neuronal arginase levels as a strategy to affect the course of TBI and provides support for the potential role of neuronal NO production in this condition.


Asunto(s)
Arginasa/genética , Lesiones Traumáticas del Encéfalo/genética , Neuronas/enzimología , Óxido Nítrico/genética , Animales , Arginina/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Línea Celular , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Humanos , Ratones , Neuronas/patología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo III/genética , Antígenos Thy-1/genética
13.
Endocrinology ; 158(3): 477-489, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27967239

RESUMEN

Bone metastasis is a deadly consequence of cancers, in which osteoclast forms a vicious cycle with tumor cells. Bone metastasis attenuation by clinical usage of osteoclast inhibitors and in our osteopetrotic mouse genetic models with ß-catenin constitutive activation or peroxisome proliferator-activated receptor γ deficiency fully support the important role of osteoclast in driving the bone metastatic niche. However, the mechanisms for this "partnership in crime" are underexplored. Here we show that osteoclasts reprogram their lipid secretion to support cancer cells. Metabolomic profiling reveals elevated prometastatic arachidonic acid (AA) but reduced antimetastatic lysophosphatidylcholines (LPCs). This shift in lipid osteoclastokines synergistically stimulates tumor cell proliferation, migration, survival, and expression of prometastatic genes. Pharmacologically, combined treatment with LPCs and BW-755C, an inhibitor of AA signaling via blocking lipoxygenase and cyclooxygenase, impedes breast cancer bone metastasis. Our findings elucidate key paracrine mechanisms for the osteoclast-cancer vicious cycle and uncover important therapeutic targets for bone metastasis.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Citocinas/metabolismo , Metabolismo de los Lípidos , Osteoclastos/fisiología , Animales , Ácido Araquidónico/metabolismo , Movimiento Celular , Femenino , Lisofosfatidilcolinas , Metabolómica , Ratones Noqueados , Ratones Desnudos , Metástasis de la Neoplasia , Trasplante de Neoplasias , Osteogénesis , Comunicación Paracrina
14.
Mol Endocrinol ; 29(5): 730-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25793404

RESUMEN

Recent studies suggest that the class II histone deacetylase (HDAC)9 plays important roles in physiology such as metabolism and immunity. Here, we report that HDAC9 also controls bone turnover by suppressing osteoclast differentiation and bone resorption. HDAC9 expression is down-regulated during osteoclastogenesis. Ex vivo osteoclast differentiation is accelerated by HDAC9 deletion but diminished by HDAC9 overexpression. HDAC9 knockout mice exhibit elevated bone resorption and lower bone mass. Bone marrow transplantation reveal that the osteoclastogenic defects are intrinsic to the hematopoietic lineage, because the excessive bone resorption phenotype can be conferred in wild-type (WT) mice receiving HDAC9-null bone marrow, and rescued in HDAC9-null mice receiving WT bone marrow. Mechanistically, HDAC9 forms a negative regulatory loop with peroxisome proliferator-activated receptor gamma (PPARg) and receptor activator of nuclear factor kappa-B ligand (RANKL) signaling. On one hand, PPARγ and nuclear factor κB suppress HDAC9 expression, on the other hand, HDAC9 inhibits PPARγ activity in synergy with silencing mediator of retinoic acid and thyroid hormone receptors (SMRT)/NCoR corepressors. These findings identify HDAC9 as a novel, important and physiologically relevant modulator of bone remodeling and skeletal homeostasis.


Asunto(s)
Histona Desacetilasas/fisiología , Osteoclastos/fisiología , PPAR gamma/metabolismo , Ligando RANK/metabolismo , Proteínas Represoras/fisiología , Animales , Remodelación Ósea , Diferenciación Celular , Regulación hacia Abajo , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
15.
Endocrinology ; 156(4): 1504-13, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25590242

RESUMEN

Adiponectin is an important adipokine. Increasing evidence suggests that altered adiponectin levels are linked with metabolic and inflammatory disorders. Here we report an important yet previously unrecognized function of adiponectin in lactation by which maternal adiponectin determines the inflammatory status in the nursing neonates. Surprisingly, both maternal adiponectin overexpression in the transgenic mice and maternal adiponectin deletion in the knockout mice lead to systemic inflammation in the pups, manifested as transient hair loss. However, distinct mechanisms are involved. Adiponectin deficiency triggers leukocyte infiltration and production of inflammatory cytokines in the lactating mammary gland. In contrast, adiponectin overabundance increases lipid accumulation in the lactating mammary gland, resulting in excessive long-chain saturated fatty acids in milk. Interestingly, in both cases, the inflammation and alopecia in the pups can be rescued by Toll-like receptor (TLR)-2/4 deletion because TLR2/4 double-knockout pups are resistant. Mechanistically, long-chain saturated fatty acid activation of inflammatory genes is TLR2/4 dependent and can be potentiated by proinflammatory cytokines, indicating that the inflammatory stimuli in both scenarios functionally converge by activating the TLR2/4 signaling. Therefore, our findings reveal adiponectin as a dosage-dependent regulator of lactation homeostasis and milk quality that critically controls inflammation in the nursing neonates. Furthermore, these results suggest that inflammatory infantile disorders may result from maternal adiponectin dysregulation that can be treated by TLR2/4 inhibition.


Asunto(s)
Adiponectina/metabolismo , Citocinas/análisis , Inflamación/prevención & control , Lactancia/metabolismo , Lípidos/análisis , Leche/química , Adiponectina/genética , Animales , Animales Recién Nacidos , Femenino , Inflamación/metabolismo , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos
16.
Mol Endocrinol ; 29(2): 172-86, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25549044

RESUMEN

Osteoclasts are bone-resorbing cells essential for skeletal remodeling and regeneration. However, excessive osteoclasts often contribute to prevalent bone degenerative diseases such as osteoporosis, arthritis, and cancer bone metastasis. Osteoclast dysregulation is also associated with rare disorders such as osteopetrosis, pycnodysostosis, Paget's disease, and Gorham-Stout syndrome. The nuclear receptor (NR) family of transcription factors functions as metabolic sensors that control a variety of physiological processes including skeletal homeostasis and serves as attractive therapeutic targets for many diseases. In this review, we highlight recent findings on the new players and the new mechanisms for how NRs regulate osteoclast differentiation and bone resorption. An enhanced understanding of NR functions in osteoclastogenesis will facilitate the development of not only novel osteoprotective medicine but also prudent strategies to minimize the adverse skeletal effects of certain NR-targeting drugs for a better treatment of cancer and metabolic diseases.


Asunto(s)
Remodelación Ósea , Osteoclastos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Humanos
17.
Cell Metab ; 20(3): 483-98, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25130399

RESUMEN

Mitochondrial complex I (CI) deficiency is associated with multiple neurological and metabolic disorders. However, its effect on innate immunity and bone remodeling is unclear. Using deletion of the essential CI subunit Ndufs4 as a model for mitochondrial dysfunction, we report that mitochondria suppress macrophage activation and inflammation while promoting osteoclast differentiation and bone resorption via both cell-autonomous and systemic regulation. Global Ndufs4 deletion causes systemic inflammation and osteopetrosis. Hematopoietic Ndufs4 deletion causes an intrinsic lineage shift from osteoclast to macrophage. Liver Ndufs4 deletion causes a metabolic shift from fatty acid oxidation to glycolysis, accumulating fatty acids and lactate (FA/LAC) in the circulation. FA/LAC further activates Ndufs4(-/-) macrophages via reactive oxygen species induction and diminishes osteoclast lineage commitment in Ndufs4(-/-) progenitors; both inflammation and osteopetrosis in Ndufs4(-/-) mice are attenuated by TLR4/2 deletion. Together, these findings reveal mitochondrial CI as a critical rheostat of innate immunity and skeletal homeostasis.


Asunto(s)
Resorción Ósea/complicaciones , Resorción Ósea/inmunología , Complejo I de Transporte de Electrón/deficiencia , Macrófagos/patología , Enfermedades Mitocondriales/complicaciones , Enfermedades Mitocondriales/inmunología , Osteoclastos/patología , Alopecia/complicaciones , Alopecia/genética , Alopecia/inmunología , Alopecia/patología , Animales , Resorción Ósea/genética , Resorción Ósea/patología , Diferenciación Celular , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/inmunología , Ácidos Grasos/metabolismo , Femenino , Eliminación de Gen , Glucólisis , Inmunidad Innata , Activación de Macrófagos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/inmunología , Mitocondrias/patología , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Osteoclastos/citología , Osteoclastos/inmunología , Especies Reactivas de Oxígeno/metabolismo
18.
Nature ; 512(7515): 431-5, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25043055

RESUMEN

Bone-resorbing osteoclasts significantly contribute to osteoporosis and bone metastases of cancer. MicroRNAs play important roles in physiology and disease, and present tremendous therapeutic potential. Nonetheless, how microRNAs regulate skeletal biology is underexplored. Here we identify miR-34a as a novel and critical suppressor of osteoclastogenesis, bone resorption and the bone metastatic niche. miR-34a is downregulated during osteoclast differentiation. Osteoclastic miR-34a-overexpressing transgenic mice exhibit lower bone resorption and higher bone mass. Conversely, miR-34a knockout and heterozygous mice exhibit elevated bone resorption and reduced bone mass. Consequently, ovariectomy-induced osteoporosis, as well as bone metastasis of breast and skin cancers, are diminished in osteoclastic miR-34a transgenic mice, and can be effectively attenuated by miR-34a nanoparticle treatment. Mechanistically, we identify transforming growth factor-ß-induced factor 2 (Tgif2) as an essential direct miR-34a target that is pro-osteoclastogenic. Tgif2 deletion reduces bone resorption and abolishes miR-34a regulation. Together, using mouse genetic, pharmacological and disease models, we reveal miR-34a as a key osteoclast suppressor and a potential therapeutic strategy to confer skeletal protection and ameliorate bone metastasis of cancers.


Asunto(s)
Neoplasias Óseas/prevención & control , Neoplasias Óseas/secundario , Diferenciación Celular/genética , MicroARNs/genética , Osteoclastos/patología , Osteoporosis/prevención & control , Proteínas Represoras/deficiencia , Animales , Secuencia de Bases , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/genética , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Neoplasias Mamarias Animales/patología , Ratones , Ratones Transgénicos , MicroARNs/farmacología , MicroARNs/uso terapéutico , Trasplante de Neoplasias , Tamaño de los Órganos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoporosis/genética , Osteoporosis/patología , Ovariectomía , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Neoplasias Cutáneas/patología , Transgenes , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cell Metab ; 19(6): 927-40, 2014 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-24794976

RESUMEN

Orexin neuropeptides promote arousal, appetite, reward, and energy expenditure. However, whether orexin affects bone mass accrual is unknown. Here, we show that orexin functions centrally through orexin receptor 2 (OX2R) in the brain to enhance bone formation. OX2R null mice exhibit low bone mass owing to elevated circulating leptin, whereas central administration of an OX2R-selective agonist augments bone mass. Conversely, orexin also functions peripherally through orexin receptor 1 (OX1R) in the bone to suppress bone formation. OX1R null mice exhibit high bone mass owing to a differentiation shift from marrow adipocyte to osteoblast that results from higher osseous ghrelin expression. The central action is dominant because bone mass is reduced in orexin null and OX1R2R double null mice but enhanced in orexin-overexpressing transgenic mice. These findings reveal orexin as a critical rheostat of skeletal homeostasis that exerts a yin-yang dual regulation and highlight orexin as a therapeutic target for osteoporosis.


Asunto(s)
Densidad Ósea/fisiología , Remodelación Ósea/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuropéptidos/metabolismo , Receptores de Orexina/metabolismo , Osteogénesis/fisiología , Animales , Densidad Ósea/genética , Diferenciación Celular/fisiología , Células Cultivadas , Ghrelina/biosíntesis , Leptina/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Receptores de Orexina/agonistas , Receptores de Orexina/genética , Orexinas , Interferencia de ARN , ARN Interferente Pequeño
20.
Mol Endocrinol ; 27(2): 325-35, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23204328

RESUMEN

The bone-resorbing osteoclast is essential for skeletal remodeling, yet its deregulation contributes to diseases such as osteoporosis and cancer bone metastasis. Here we identify histone deacetylase 7 (HDAC7) as a key negative regulator of osteoclastogenesis and bone resorption using both in vitro cellular and molecular analyses and in vivo characterization of conditional HDAC7-knockout mice. Bone marrow osteoclast differentiation assays reveal that HDAC7 overexpression suppresses, whereas HDAC7 deletion enhances, osteoclastogenesis. Mechanistically, in the absence of receptor activator of nuclear factor κ-B ligand (RANKL), HDAC7 attenuates ß-catenin function and cyclin D1 expression, thereby reducing precursor proliferation; upon RANKL activation, HDAC7 suppresses NFATc1 and prevents ß-catenin down-regulation, thereby blocking osteoclast differentiation. Consequently, HDAC7 deletion in the osteoclast lineage results in a 26% reduction in bone mass (P = 0.003) owing to 102% elevated bone resorption (P = 0.01). These findings are clinically significant in light of the remarkable therapeutic potentials of HDAC inhibitors for several diseases such as cancer, diabetes, and neurodegeneration.


Asunto(s)
Histona Desacetilasas/metabolismo , Osteoclastos/metabolismo , Ligando RANK/metabolismo , beta Catenina/metabolismo , Animales , Densidad Ósea , Remodelación Ósea , Resorción Ósea/genética , Resorción Ósea/metabolismo , Huesos/metabolismo , Diferenciación Celular , Línea Celular , Proliferación Celular , Ciclina D1/biosíntesis , Ciclina D1/metabolismo , Regulación hacia Abajo , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , Osteoporosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...