Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Lipid Res ; 63(12): 100297, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36243101

RESUMEN

Bile acids (BAs) are steroid detergents in bile that contribute to fat absorption, cell signaling, and microbiome interactions. The final step in their synthesis is amino acid conjugation with either glycine or taurine in the liver by the enzyme bile acid-CoA:amino acid N-acyltransferase (BAAT). Here, we describe the microbial, chemical, and physiological consequences of Baat gene knockout. Baat-/- mice were underweight after weaning but quickly exhibited catch-up growth. At three weeks of age, KO animals had increased phospholipid excretion and decreased subcutaneous fat pad mass, liver mass, glycogen staining in hepatocytes, and hepatic vitamin A stores, but these were less marked in adulthood. Additionally, KO mice had an altered microbiome in early life. Their BA pool was highly enriched in cholic acid but not completely devoid of conjugated BAs. KO animals had 27-fold lower taurine-conjugated BAs than wild type in their liver but similar concentrations of glycine-conjugated BAs and higher microbially conjugated BAs. Furthermore, the BA pool in Baat-/- was enriched in a variety of unusual BAs that were putatively sourced from cysteamine conjugation with subsequent oxidation and methylation of the sulfur group mimicking taurine. Antibiotic treatment of KO mice indicated the microbiome was not the likely source of the unusual conjugations, instead, the unique BAs in KO animals were likely derived from the peroxisomal acyltransferases Acnat1 and Acnat2, which are duplications of Baat in the mouse genome that are inactivated in humans. This study demonstrates that BA conjugation is important for early life development of mice.


Asunto(s)
Ácidos y Sales Biliares , Microbioma Gastrointestinal , Humanos , Ratones , Animales , Adulto , Técnicas de Inactivación de Genes , Ratones Noqueados , Hígado/metabolismo , Taurina/metabolismo , Glicina
2.
Am J Physiol Endocrinol Metab ; 323(2): E159-E170, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35658543

RESUMEN

Undernutrition-induced growth restriction in the early stages of life increases the risk of chronic disease in adulthood. Although metabolic impairments have been observed, few studies have characterized the gut microbiome and gut-liver metabolome profiles of growth-restricted animals during early-to-mid-life development. To induce growth restriction, mouse offspring were either born to gestational undernutrition (GUN) or suckled from postnatal undernutrition (PUN) dams fed a protein-restricted diet (8% protein) or control diet (CON; 20% protein) until weaning at postnatal age of 21 days (PN21). At PN21, all mice were fed the CON diet until adulthood (PN80). Livers were collected at PN21 and PN80, and fecal samples were collected weekly starting at PN21 (postweaning week 1) until PN80 (postweaning week 5) for gut microbiome and metabolome analyses. PUN mice exhibited the most alterations in gut microbiome and gut and liver metabolome compared with CON mice. These mice had altered fecal microbial ß-diversity (P = 0.001) and exhibited higher proportions of Bifidobacteriales [linear mixed model (LMM) P = 7.1 × 10-6), Clostridiales (P = 1.459 × 10-5), Erysipelotrichales (P = 0.0003), and lower Bacteroidales (P = 4.1 × 10-5)]. PUN liver and fecal metabolome had a reduced total bile acid pool (P < 0.01), as well as lower abundance of riboflavin (P = 0.003), amino acids [i.e., methionine (P = 0.0018), phenylalanine (P = 0.0015), and tyrosine (P = 0.0041)], and higher excreted total peptides (LMM P = 0.0064) compared with CON. Overall, protein restriction during lactation permanently alters the gut microbiome into adulthood. Although the liver bile acids, amino acids, and acyl-carnitines recovered, the fecal peptides and microbiome remained permanently altered into adulthood, indicating that inadequate protein intake in a specific time frame in early life can have an irreversible impact on the microbiome and fecal metabolome.NEW & NOTEWORTHY Undernutrition-induced early-life growth restriction not only leads to increased disease risk but also permanently alters the gut microbiome and gut-liver metabolome during specific windows of early-life development.


Asunto(s)
Microbioma Gastrointestinal , Desnutrición , Animales , Ácidos y Sales Biliares , Dieta con Restricción de Proteínas , Heces , Femenino , Metaboloma , Ratones
3.
J Cell Physiol ; 235(3): 2350-2365, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31538345

RESUMEN

Type-1 diabetes (T1D) increases systemic inflammation, bone loss, and risk for bone fractures. Levels of the anti-inflammatory cytokine interleukin-10 (IL-10) are decreased in T1D, however their role in T1D-induced osteoporosis is unknown. To address this, diabetes was induced in male IL-10 knockout (KO) and wild-type (WT) mice. Analyses of femur and vertebral trabecular bone volume fraction identified bone loss in T1D-WT mice at 4 and 12 weeks, which in T1D-IL-10-KO mice was further reduced at 4 weeks but not 12 weeks. IL-10 deficiency also increased the negative effects of T1D on cortical bone. Osteoblast marker osterix was decreased, while osteoclast markers were unchanged, suggesting that IL-10 promotes anabolic processes. MC3T3-E1 osteoblasts cultured under high glucose conditions displayed a decrease in osterix which was prevented by addition of IL-10. Taken together, our results suggest that IL-10 is important for promoting osteoblast maturation and reducing bone loss during early stages of T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Fracturas Óseas/genética , Interleucina-10/genética , Osteoporosis/genética , Factor de Transcripción Sp7/genética , Animales , Hueso Esponjoso/metabolismo , Hueso Esponjoso/patología , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/patología , Fémur/metabolismo , Fémur/patología , Fracturas Óseas/complicaciones , Fracturas Óseas/patología , Glucosa/metabolismo , Humanos , Inflamación/complicaciones , Inflamación/genética , Inflamación/patología , Ratones Noqueados , Osteoblastos/metabolismo , Osteoblastos/patología , Osteoclastos/metabolismo , Osteoclastos/patología , Osteoporosis/complicaciones , Osteoporosis/patología , Factores de Riesgo
4.
J Cell Biochem ; 120(3): 4398-4408, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30269370

RESUMEN

Leptin, a hormone primarily produced by adipocytes, contributes to the regulation of bone health by modulating bone density, growth and adiposity. Upon leptin binding, multiple sites of the long form of the leptin receptor (LepRb) are phosphorylated to trigger activation of downstream signaling pathways. To address the role of LepRb-signaling pathways in bone health, we compared the effects of three LepRb mutations on bone density, adiposity, and growth in male and female mice. The ∆65 mutation, which lacks the known tyrosine phosphorylation sites, caused obesity and the most dramatic bone phenotype marked by excessive bone adiposity, osteoporosis, and decreased growth, consistent with the phenotype of db/db and ob/ob mice that fully lack leptin receptor signaling. Mutation of LepRb Tyr 1138 , which results in an inability to recruit and phosphorylate signal transducer and activator of transcription 3, also caused obesity, but bone loss and adiposity were more dominant in male mice and no growth defect was observed. In contrast, mutation of LepRb Tyr 985 , which blocks SHP2/SOCS3 recruitment to LepRb and contributes to leptin hypersensitivity, promoted increased femur bone density only in male mice, while marrow adiposity and bone growth were not affected. Additional analyses of vertebral trabecular bone volume indicate that only the Tyr 1138 mutant mice exhibit bone loss in vertebrae. Together, our findings suggest that the phosphorylation status of specific sites of the LepRb contribute to the sex- and location-dependent bone responses to leptin. Unraveling the mechanisms by which leptin responses are sex- and location-dependent can contribute to the development of uniquely targeted osteoporosis therapies.


Asunto(s)
Adiposidad/fisiología , Densidad Ósea/fisiología , Leptina/metabolismo , Receptores de Leptina/metabolismo , Caracteres Sexuales , Transducción de Señal/fisiología , Adipocitos Blancos/metabolismo , Animales , Hueso Esponjoso/metabolismo , Femenino , Fémur/metabolismo , Leptina/genética , Masculino , Ratones , Ratones Mutantes , Mutación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Receptores de Leptina/genética , Columna Vertebral/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo
5.
BMC Musculoskelet Disord ; 20(1): 326, 2019 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-31299941

RESUMEN

BACKGROUND: Oestrogen-deficiency induced by menopause is associated with reduced bone density and primary osteoporosis, resulting in an increased risk of fracture. While the exact etiology of menopause-induced primary osteoporotic bone loss is not fully known, members of the tumour necrosis factor super family (TNFSF) are known to play a role. Recent studies have revealed that the TNFSF members death receptor 3 (DR3) and one of its ligands, TNF-like protein 1A (TL1A) have a key role in secondary osteoporosis; enhancing CD14+ peripheral blood mononuclear cell (PBMC) osteoclast formation and bone resorption. Whether DR3 and TL1A contribute towards bone loss in menopause-induced primary osteoporosis however, remains unknown. METHODS: To investigate this we performed flow cytometry analysis of DR3 expression on CD14+ PBMCs isolated from pre- and early post-menopausal females and late post-menopausal osteoporotic patients. Serum levels of TL1A, CCL3 and total MMP-9 were measured by ELISA. In vitro osteoclast differentiation assays were performed to determine CD14+ monocyte osteoclastogenic potential. In addition, splenic CD4+ T cell DR3 expression was investigated 1 week and 8 weeks post-surgery, using the murine ovariectomy model. RESULTS: In contrast to pre-menopausal females, CD14+ monocytes isolated from post-menopausal females were unable to induce DR3 expression. Serum TL1A levels were decreased approx. 2-fold in early post-menopausal females compared to pre-menopausal controls and post-menopausal osteoporotic females; no difference was observed between pre-menopausal and late post-menopausal osteoporotic females. Analysis of in vitro CD14+ monocyte osteoclastogenic potential revealed no significant difference between the post-menopausal and post-menopausal osteoporotic cohorts. Interestingly, in the murine ovariectomy model splenic CD4+ T cell DR3 expression was significantly increased at 1 week but not 8 weeks post-surgery when compared to the sham control. CONCLUSION: Our results reveals for the first time that loss of oestrogen has a significant effect on DR3; decreasing expression on CD14+ monocytes and increasing expression on CD4+ T cells. These data suggest that while oestrogen-deficiency induced changes in DR3 expression do not affect late post-menopausal bone loss they could potentially have an indirect role in early menopausal bone loss through the modulation of T cell activity.


Asunto(s)
Estrógenos/deficiencia , Osteoporosis Posmenopáusica/metabolismo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/sangre , Adulto , Anciano , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Receptores de Lipopolisacáridos/metabolismo , Menopausia/sangre , Menopausia/fisiología , Ratones , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Osteoporosis Posmenopáusica/sangre , Osteoporosis Posmenopáusica/inmunología , Ovariectomía , Adulto Joven
6.
Physiol Genomics ; 50(6): 407-415, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29570431

RESUMEN

G protein-coupled receptor kinase 2 (GRK2) is a serine/threonine kinase and plays a key role in different disease processes. Previously, we showed that GRK2 knockdown enhances wound healing in colonic epithelial cells. Therefore, we hypothesized that ablation of GRK2 would protect mice from dextran sodium sulfate (DSS)-induced acute colitis. To test this, we administered DSS to wild-type (GRK2+/+) and GRK2 heterozygous (GRK+/-) mice in their drinking water for 7 days. As predicted, GRK2+/- mice were protected from colitis as demonstrated by decreased weight loss (20% loss in GRK2+/+ vs. 11% loss in GRK2+/-). lower disease activity index (GRK2+/+ 9.1 vs GRK2+/- 4.1), and increased colon lengths (GRK2+/+ 4.7 cm vs GRK2+/- 5.3 cm). To examine the mechanisms by which GRK2+/- mice are protected from colitis, we investigated expression of inflammatory genes in the colon as well as immune cell profiles in colonic lamina propria, mesenteric lymph node, and in bone marrow. Our results did not reveal differences in immune cell profiles between the two genotypes. However, expression of inflammatory genes was significantly decreased in DSS-treated GRK2+/- mice compared with GRK2+/+. To understand the mechanisms, we generated myeloid-specific GRK2 knockout mice and subjected them to DSS-induced colitis. Similar to whole body GRK2 heterozygous knockout mice, myeloid-specific knockout of GRK2 was sufficient for the protection from DSS-induced colitis. Together our results indicate that deficiency of GRK2 protects mice from DSS-induced colitis and further suggests that the mechanism of this effect is likely via GRK2 regulation of inflammatory genes in the myeloid cells.


Asunto(s)
Colitis/inducido químicamente , Colitis/prevención & control , Quinasa 2 del Receptor Acoplado a Proteína-G/deficiencia , Enfermedad Aguda , Animales , Colitis/enzimología , Colitis/patología , Colon/metabolismo , Colon/patología , Sulfato de Dextran , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Heterocigoto , Inflamación/genética , Inflamación/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo
7.
Toxicol Appl Pharmacol ; 348: 85-98, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29673856

RESUMEN

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and other aryl hydrocarbon receptor (AhR) agonists have been shown to regulate bone development and remodeling in a species-, ligand-, and age-specific manner, however the underlying mechanisms remain poorly understood. In this study, we characterized the effect of 0.01-30 µg/kg TCDD on the femoral morphology of male and female juvenile mice orally gavaged every 4 days for 28 days and used RNA-Seq to investigate gene expression changes associated with the resultant phenotype. Micro-computed tomography revealed that TCDD dose-dependently increased trabecular bone volume fraction (BVF) 2.9- and 3.3-fold in male and female femurs, respectively. Decreased serum tartrate-resistant acid phosphatase (TRAP) levels, combined with a reduced osteoclast surface to bone surface ratio and repression of femoral proteases (cathepsin K, matrix metallopeptidase 13), suggests that TCDD impaired bone resorption. Increased osteoblast counts at the trabecular bone surface were consistent with a reciprocal reduction in the number of bone marrow adipocytes, suggesting AhR activation may direct mesenchymal stem cell differentiation towards osteoblasts rather than adipocytes. Notably, femoral expression of transmembrane glycoprotein NMB (Gpnmb; osteoactivin), a positive regulator of osteoblast differentiation and mineralization, was dose-dependently induced up to 18.8-fold by TCDD. Moreover, increased serum levels of 1,25-dihydroxyvitamin D3 were in accordance with the renal induction of 1α-hydroxylase Cyp27b1 and may contribute to impaired bone resorption. Collectively, the data suggest AhR activation tipped the bone remodeling balance towards bone formation, resulting in increased bone mass with reduced marrow adiposity.


Asunto(s)
Adiposidad/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Hueso Esponjoso/efectos de los fármacos , Fémur/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/biosíntesis , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Factores de Edad , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Médula Ósea/metabolismo , Médula Ósea/fisiopatología , Resorción Ósea/inducido químicamente , Resorción Ósea/metabolismo , Resorción Ósea/fisiopatología , Calcitriol/sangre , Hueso Esponjoso/diagnóstico por imagen , Hueso Esponjoso/metabolismo , Hueso Esponjoso/fisiopatología , Catepsina K/metabolismo , Relación Dosis-Respuesta a Droga , Proteínas del Ojo/metabolismo , Femenino , Fémur/diagnóstico por imagen , Fémur/metabolismo , Fémur/fisiopatología , Riñón/enzimología , Masculino , Metaloproteinasa 13 de la Matriz/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos C57BL , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/metabolismo , Fosfatasa Ácida Tartratorresistente/sangre , Factores de Tiempo , Microtomografía por Rayos X
8.
Calcif Tissue Int ; 102(4): 480-488, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29453726

RESUMEN

Probiotics have been consumed by humans for thousands of years because they are beneficial for long-term storage of foods and promote the health of their host. Ingested probiotics reside in the gastrointestinal tract where they have many effects including modifying the microbiota composition, intestinal barrier function, and the immune system which result in systemic benefits to the host, including bone health. Probiotics benefit bone growth, density, and structure under conditions of dysbiosis, intestinal permeability, and inflammation (recognized mediators of bone loss and osteoporosis). It is likely that multiple mechanisms are involved in mediating probiotic signals from the gut to the bone. Studies indicate a role for the microbiota (composition and activity), intestinal barrier function, and immune cells in the signaling process. These mechanisms are not mutually exclusive, but rather, may synergize to provide benefits to the skeletal system of the host and serve as a starting point for investigation. Given that probiotics hold great promise for supporting bone health and are generally regarded as safe, future studies identifying mechanisms are warranted.


Asunto(s)
Huesos/inmunología , Microbiota/inmunología , Minerales/metabolismo , Osteoporosis/microbiología , Probióticos/metabolismo , Animales , Humanos , Inflamación/inmunología , Inflamación/microbiología , Osteoporosis/inmunología
9.
Biochem J ; 474(14): 2301-2313, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28572156

RESUMEN

G-protein-coupled receptor kinase-2 (GRK2) belongs to the GRK family of serine/threonine protein kinases critical in the regulation of G-protein-coupled receptors. Apart from this canonical role, GRK2 is also involved in several signaling pathways via distinct intracellular interactomes. In the present study, we examined the role of GRK2 in TNFα signaling in colon epithelial cell-biological processes including wound healing, proliferation, apoptosis, and gene expression. Knockdown of GRK2 in the SW480 human colonic cells significantly enhanced TNFα-induced epithelial cell wound healing without any effect on apoptosis/proliferation. Consistent with wound-healing effects, GRK2 knockdown augmented TNFα-induced matrix metalloproteinases (MMPs) 7 and 9, as well as urokinase plasminogen activator (uPA; factors involved in cell migration and wound healing). To assess the mechanism by which GRK2 affects these physiological processes, we examined the role of GRK2 in TNFα-induced MAPK and NF-κB pathways. Our results demonstrate that while GRK2 knockdown inhibited TNFα-induced IκBα phosphorylation, activation of ERK was significantly enhanced in GRK2 knockdown cells. Our results further demonstrate that GRK2 inhibits TNFα-induced ERK activation by inhibiting generation of reactive oxygen species (ROS). Together, these data suggest that GRK2 plays a critical role in TNFα-induced wound healing by modulating MMP7 and 9 and uPA levels via the ROS-ERK pathway. Consistent with in vitro findings, GRK2 heterozygous mice exhibited enhanced intestinal wound healing. Together, our results identify a novel role for GRK2 in TNFα signaling in intestinal epithelial cells.


Asunto(s)
Colon/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Mucosa Intestinal/metabolismo , Sistema de Señalización de MAP Quinasas , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Colon/citología , Colon/inmunología , Quinasa 2 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Regulación Enzimológica de la Expresión Génica , Heterocigoto , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Metaloproteinasas de la Matriz Secretadas/genética , Metaloproteinasas de la Matriz Secretadas/metabolismo , Ratones , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/metabolismo , Organismos Libres de Patógenos Específicos , Factor de Necrosis Tumoral alfa/genética , Cicatrización de Heridas
10.
Physiol Genomics ; 49(11): 682-689, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28939643

RESUMEN

G protein-coupled receptor kinase-6 (GRK6) is a serine/threonine kinase that is important in inflammatory processes. In this study, we examined the role of GRK6 in Escherichia coli-induced lung infection and inflammation using GRK6 knockout (KO) and wild-type (WT) mice. Intratracheal instillation of E. coli significantly enhanced bacterial load in the bronchoalveolar lavage (BAL) of KO compared with WT mice. Reduced bacterial clearance in the KO mice was not due to an intrinsic defect in neutrophil phagocytosis or killing but as a result of reduced neutrophil numbers in the KO BAL. Interestingly, neutrophil numbers in the lung were increased in the KO compared with WT mice, suggesting a potential dysfunction in transepithelial migration of neutrophils from the lungs to the bronchoalveolar space. This effect was selective for lung tissue because peritoneal neutrophil numbers were similar between the two genotypes following peritoneal infection. Although neutrophil expression of CXCR2/CXCR3 was similar between WT and KO, IL-17A expression was higher in the KO compared with WT mice. These results suggest that enhanced neutrophil count in the KO lungs but reduced numbers in BAL are likely due to transepithelial migration defect and/or altered chemokines/cytokines. Together, our studies suggest a previously unrecognized and novel role for GRK6 in neutrophil migration specific to pulmonary tissue during bacterial infection.


Asunto(s)
Infecciones por Escherichia coli/enzimología , Infecciones por Escherichia coli/microbiología , Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Enfermedades Pulmonares/enzimología , Enfermedades Pulmonares/microbiología , Animales , Apoptosis/genética , Carga Bacteriana , Líquido del Lavado Bronquioalveolar/citología , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/patología , Regulación de la Expresión Génica , Inflamación/genética , Inflamación/patología , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Viabilidad Microbiana , Neutrófilos/metabolismo , Fagocitosis , Receptores de Quimiocina/metabolismo
11.
Adv Exp Med Biol ; 1033: 225-247, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29101658

RESUMEN

The intestinal environment is linked to an array of conditions and diseases, including osteoporosis. Human and animal studies indicate that probiotics can benefit intestinal health and may provide a useful therapeutic to prevent and/or treat bone loss. Probiotics are defined as live microorganisms that when administered in adequate amounts will confer a health benefit on the host. In this review, we will focus on (1) probiotics (definition, history, nomenclature, types), (2) the effects of probiotics on bone health, and (3) mechanisms of probiotic prevention of bone pathologies.


Asunto(s)
Huesos/fisiología , Tracto Gastrointestinal/fisiología , Probióticos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Animales , Enfermedades Óseas/fisiopatología , Enfermedades Óseas/prevención & control , Humanos , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Probióticos/administración & dosificación
12.
Adv Exp Med Biol ; 1033: 151-183, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29101655

RESUMEN

The intestinal epithelial barrier plays an essential role in maintaining host homeostasis. The barrier regulates nutrient absorption as well as prevents the invasion of pathogenic bacteria in the host. It is composed of epithelial cells, tight junctions, and a mucus layer. Several factors, such as cytokines, diet, and diseases, can affect this barrier. These factors have been shown to increase intestinal permeability, inflammation, and translocation of pathogenic bacteria. In addition, dysregulation of the epithelial barrier can result in inflammatory diseases such as inflammatory bowel disease. Our lab and others have also shown that barrier disruption can have systemic effects including bone loss. In this chapter, we will discuss the current literature to understand the link between intestinal barrier and bone. We will discuss how inflammation, aging, dysbiosis, and metabolic diseases can affect intestinal barrier-bone link. In addition, we will highlight the current suggested mechanism between intestinal barrier and bone.


Asunto(s)
Huesos/fisiología , Tracto Gastrointestinal/fisiología , Mucosa Intestinal/fisiología , Transducción de Señal , Uniones Estrechas/fisiología , Animales , Disbiosis/fisiopatología , Humanos , Inflamación/fisiopatología , Mucosa Intestinal/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/metabolismo
13.
Adv Exp Med Biol ; 1033: 59-94, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29101652

RESUMEN

In recent years a link between the gastrointestinal tract and bone health has started to gain significant attention. Dysbiosis of the intestinal microbiota has been linked to the pathology of a number of diseases which are associated with bone loss. In addition modulation of the intestinal microbiota with probiotic bacteria has revealed to have both beneficial local and systemic effects. In the present chapter, we discuss the intestinal and bone immune systems, explore how intestinal disease affects the immune system, and examine how these pathologic changes could adversely impact bone health.


Asunto(s)
Huesos/inmunología , Tracto Gastrointestinal/inmunología , Sistema Inmunológico/inmunología , Transducción de Señal/inmunología , Animales , Remodelación Ósea/inmunología , Huesos/citología , Huesos/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/metabolismo , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/metabolismo , Enfermedades Intestinales/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo
14.
Am J Physiol Regul Integr Comp Physiol ; 311(6): R1149-R1157, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27733383

RESUMEN

Increasing evidence indicates a strong link between intestinal health and bone health. For example, inflammatory bowel disease can cause systemic inflammation, weight loss, and extra-intestinal manifestations, such as decreased bone growth and density. However, the effects of moderate intestinal inflammation without weight loss on bone health have never been directly examined; yet this condition is relevant not only to IBD but to conditions of increased intestinal permeability and inflammation, as seen with ingestion of high-fat diets, intestinal dysbiosis, irritable bowel syndrome, metabolic syndrome, and food allergies. Here, we induced moderate intestinal inflammation without weight loss in young male mice by treating with a low dose of dextran sodium sulfate (1%) for 15 days. The mice displayed systemic changes marked by significant bone loss and a redistribution of fat from subcutaneous to visceral fat pad stores. Bone loss was caused by reduced osteoblast activity, characterized by decreased expression of osteoblast markers (runx2, osteocalcin), histomorphometry, and dynamic measures of bone formation. In addition, we observed a reduction in growth plate thickness and hypertrophic chondrocyte matrix components (collagen X). Correlation analyses indicate a link between gut inflammation and disease score, but more importantly, we observed that bone density measures negatively correlated with intestinal disease score, as well as colon and bone TNF-α levels. These studies demonstrate that colitis-induced bone loss is not dependent upon weight loss and support a role for inflammation in the link between gut and bone health, an important area for future therapeutic development.


Asunto(s)
Tejido Adiposo/fisiopatología , Densidad Ósea , Desarrollo Óseo , Colitis/fisiopatología , Osteoporosis/fisiopatología , Pérdida de Peso , Animales , Colitis/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoporosis/etiología , Tibia/fisiopatología
15.
J Cell Physiol ; 230(8): 1944-53, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25641511

RESUMEN

Type I (T1) diabetes is an autoimmune and metabolic disease associated with bone loss. Previous studies demonstrate that T1-diabetes decreases osteoblast activity and viability. Bisphosphonate therapy, commonly used to treat osteoporosis, is demonstrated to inhibit osteoclast activity as well as osteoblast apoptosis. Therefore, we examined the effect of weekly alendronate treatments on T1-diabetes induced osteoblast apoptosis and bone loss. Bone TUNEL assays identified that alendronate therapy prevents the diabetes-induced osteoblast death observed during early stages of diabetes development. Consistent with this, alendronate treatment for 40 days was able to prevent diabetes-induced trabecular bone loss. Alendronate was also able to reduce marrow adiposity in both control diabetic mice compared to untreated mice. Mechanical testing indicated that 40 days of alendronate treatment increased bone stiffness but decreased the work required for fracture in T1-diabetic and alendronate treated mice. Of concern at this later time point, bone formation rate and osteoblast markers, which were already decreased in diabetic mice, were further suppressed in alendronate-treated diabetic mice. Taken together, our results suggest that short-term alendronate treatment can prevent T1-diabetes-induced bone loss in mice, possibly in part by inhibiting diabetes onset associated osteoblast death, while longer treatment enhanced bone density but at the cost of further suppressing bone formation in diabetic mice.


Asunto(s)
Alendronato/toxicidad , Conservadores de la Densidad Ósea/toxicidad , Resorción Ósea , Diabetes Mellitus Tipo 1/complicaciones , Osteogénesis/efectos de los fármacos , Animales , Densidad Ósea/efectos de los fármacos , Diabetes Mellitus Experimental/complicaciones , Difosfonatos/toxicidad , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/efectos de los fármacos , Microtomografía por Rayos X
16.
Proc Natl Acad Sci U S A ; 109(41): 16594-9, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-23012474

RESUMEN

The bone marrow (BM) is a large, highly active, and responsive tissue. Interestingly, little is known about the impact of colitis on hematopoietic functions. Using dextran sodium sulfate (DSS) to induce colitis in mice, we identified significant changes in the BM. Specifically, cells of the monocytic and granulocytic lineages increased nearly 60% and 80%, respectively. This change would support and promote the large infiltration of the gut with neutrophils and monocytes that are the primary cause of inflammation and tissue damage during colitis. Conversely, the early lineages of B and T cells declined in the marrow and thymus with particularly large losses observed among pre-B and pre-T cells with heightened levels of apoptosis noted among CD4(+)CD8(+) thymocytes from DSS-treated mice. Also noteworthy was the 40% decline in cells of the erythrocytic lineages in the marrow of colitis mice, which undoubtedly contributed to the anemia observed in these mice. The peripheral blood reflected the marrow changes as demonstrated by a 2.6-fold increase in neutrophils, a 60% increase in monocytes, and a decline in the lymphocyte population. Thus, colitis changed the BM in profound ways that parallel the general outcomes of colitis including infiltration of the gut with monocytes and neutrophils, inflammation, and anemia. The data provide important understandings of the full impact of colitis that may lead to unique treatments and therapies.


Asunto(s)
Linaje de la Célula/inmunología , Colitis/inmunología , Granulocitos/inmunología , Monocitos/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Antígenos CD4/inmunología , Antígenos CD8/inmunología , Colitis/inducido químicamente , Colitis/patología , Sulfato de Dextran , Citometría de Flujo , Granulocitos/patología , Ratones , Ratones Endogámicos C57BL , Monocitos/patología , Neutrófilos/inmunología , Neutrófilos/patología , Linfocitos T/inmunología , Linfocitos T/patología , Timocitos/inmunología , Timocitos/patología , Timo/inmunología , Timo/patología , Factores de Tiempo
17.
J Cell Physiol ; 229(11): 1822-30, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24677054

RESUMEN

Estrogen deficiency is a major risk factor for osteoporosis that is associated with bone inflammation and resorption. Half of women over the age of 50 will experience an osteoporosis related fracture in their lifetime, thus novel therapies are needed to combat post-menopausal bone loss. Recent studies suggest an important role for gut-bone signaling pathways and the microbiota in regulating bone health. Given that the bacterium Lactobacillus reuteri ATCC PTA 6475 (L. reuteri) secretes beneficial immunomodulatory factors, we examined if this candidate probiotic could reduce bone loss associated with estrogen deficiency in an ovariectomized (Ovx) mouse menopausal model. Strikingly, L. reuteri treatment significantly protected Ovx mice from bone loss. Osteoclast bone resorption markers and activators (Trap5 and RANKL) as well as osteoclastogenesis are significantly decreased in L. reuteri-treated mice. Consistent with this, L. reuteri suppressed Ovx-induced increases in bone marrow CD4+ T-lymphocytes (which promote osteoclastogenesis) and directly suppressed osteoclastogenesis in vitro. We also identified that L. reuteri treatment modifies microbial communities in the Ovx mouse gut. Together, our studies demonstrate that L. reuteri treatment suppresses bone resorption and loss associated with estrogen deficiency. Thus, L. reuteri treatment may be a straightforward and cost-effective approach to reduce post-menopausal bone loss.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Resorción Ósea/prevención & control , Ovariectomía , Probióticos/uso terapéutico , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Diferenciación Celular , Línea Celular , Modelos Animales de Enfermedad , Femenino , Fémur/patología , Citometría de Flujo , Lactobacillus , Menopausia , Ratones , Ratones Endogámicos BALB C , Tamaño de los Órganos , Osteoblastos/metabolismo , Osteoblastos/patología , Osteoclastos/metabolismo , Osteoclastos/patología , Análisis de Componente Principal , Ligando RANK/metabolismo , Columna Vertebral/patología
18.
Am J Pathol ; 182(4): 1114-23, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23395087

RESUMEN

ß-Arrestins are intracellular scaffolding proteins that modulate specific cell signaling pathways. Recent studies, in both cell culture and in vivo models, have demonstrated an important role for ß-arrestin-1 in inflammation. However, the role of ß-arrestin-1 in the pathogenesis of inflammatory bowel disease (IBD) is not known. Our goal was to investigate the role of ß-arrestin-1 in IBD using mouse models of colitis. To this end, we subjected wild-type (WT) and ß-arrestin-1 knockout (ß-arr-1(-/-)) mice to colitis induced by trinitrobenzenesulfonic acid or dextran sulfate sodium and examined the clinical signs, gross pathology, and histopathology of the colon, as well as inflammatory components. The ß-arr-1(-/-) mice displayed significantly attenuated colitis, compared with WT mice, in both models. Consistent with the phenotypic observations, histological examination of the colon revealed attenuated disease pathology in the ß-arr-1(-/-) mice. Our results further demonstrate that ß-arr-1(-/-) mice are deficient in IL-6 expression in the colon, but have higher expression of the anti-inflammatory IL-10 family of cytokines. Our results also demonstrate diminished ERK and NFκB pathways in the colons of ß-arr-1(-/-) mice, compared with WT mice. Taken together, our results demonstrate that decreased IL-6 production and enhanced IL-10 and IL-22 production in ß-arrestin-1-deficient mice likely lead to attenuated gut inflammation.


Asunto(s)
Arrestinas/deficiencia , Colitis/patología , Colitis/prevención & control , Animales , Arrestinas/metabolismo , Colitis/sangre , Colitis/enzimología , Colon/patología , Sulfato de Dextran , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Inflamación/patología , Interleucina-10/sangre , Interleucina-6/sangre , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Transducción de Señal , Ácido Trinitrobencenosulfónico , Pérdida de Peso , beta-Arrestina 1 , beta-Arrestinas
19.
Front Pharmacol ; 15: 1268134, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38533264

RESUMEN

The gut microbiota and barrier function play important roles in bone health. We previously demonstrated that chronic glucocorticoid (GC)-induced bone loss in mice is associated with significant shifts in gut microbiota composition and impaired gut barrier function. Korean Red Ginseng (KRG, Panax Ginseng Meyer, Araliaceae) extract has been shown to prevent glucocorticoid-induced osteoporosis (GIO) in a subcutaneous pellet model in mice, but its effect on gut microbiota and barrier function in this context is not known. The overall goal of this study was to test the effect of KRG extract in a clinically relevant, oral model of GIO and further investigate its role in modulating the gut-bone axis. Growing male mice (CD-1, 8 weeks) were treated with 75 µg/mL corticosterone (∼9 mg/kg/day) or 0.4% ethanol vehicle in the drinking water for 4 weeks. During this 4-week period, mice were treated daily with 500 mg/kg/day KRG extract dissolved in sterile water or an equal amount of sterile water via oral gastric gavage. After 4 weeks of treatment, we assessed bone volume, microbiota composition, gut barrier integrity, and immune cells in the bone marrow (BM) and mesenteric lymph nodes (MLNs). 4 weeks of oral GC treatment caused significant distal femur trabecular bone loss, and this was associated with changes in gut microbiota composition, impaired gut barrier function and altered immune cell composition. Importantly, KRG extract prevented distal femur trabecular bone loss and caused significant alterations in gut microbiota composition but had only modest effects on gut barrier function and immune cell populations. Taken together, these results demonstrate that KRG extract significantly modulates the gut microbiota-bone axis and prevents glucocorticoid-induced bone loss in mice.

20.
J Cell Physiol ; 228(4): 689-95, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22886636

RESUMEN

The adolescent skeleton undergoes accelerated growth determining overall bone density, length, and quality. Diseases such as type 1 diabetes (T1D), most often diagnosed in adolescents, can alter bone processes and promote bone loss. Studies examining type 1 diabetic (T1D) bone pathologies typically utilize adult mice and rely on pharmacologic models such as streptozotocin (STZ)-induced diabetic rodents. To test the effect of T1D on adolescent bone growth/density we used a novel juvenile genetic model (Ins2(+/-) mice) that spontaneously develop T1D at approximately 5 weeks of age and compared our findings with STZ-induced T1D mice. Compared to controls, both Ins2(+/-) and STZ-induced T1D mice displayed blood glucose levels greater than 300 mg/dl and reduced body, fat and muscle mass as well as femur trabecular bone density. STZ mice exhibited greater bone loss compared to Ins2(+/-) mice despite having lower blood glucose levels. Cortical bone was affected in STZ but not Ins2(+/-) mice. Osteocalcin serum protein and bone RNA levels decreased in both models. Consistent with studies in adult mice, STZ adolescent mice displayed increased marrow adiposity, however this was not observed in the Ins2(+/-) mice. Reduced femur length, decreased growth plate thickness and decreased collagen II expression in both model simplies impaired cartilage formation. In summary, both pharmacologic and spontaneous adolescent T1D mice demonstrated a bone synthesis and growth defect. STZ appears to cause a more severe phenotype. Thus, the Ins2(+/-) mouse could serve as a useful model to study adolescent T1D bone loss with fewer complications.


Asunto(s)
Resorción Ósea/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Insulina/metabolismo , Animales , Glucemia/metabolismo , Densidad Ósea/fisiología , Resorción Ósea/genética , Resorción Ósea/patología , Cartílago/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animales de Enfermedad , Fémur/metabolismo , Fémur/fisiopatología , Insulina/deficiencia , Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogénesis/genética , Osteogénesis/fisiología , ARN/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA