Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Metab ; 36(3): 575-597.e7, 2024 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-38237602

RESUMEN

The glucagon receptor (GCGR) in the kidney is expressed in nephron tubules. In humans and animal models with chronic kidney disease, renal GCGR expression is reduced. However, the role of kidney GCGR in normal renal function and in disease development has not been addressed. Here, we examined its role by analyzing mice with constitutive or conditional kidney-specific loss of the Gcgr. Adult renal Gcgr knockout mice exhibit metabolic dysregulation and a functional impairment of the kidneys. These mice exhibit hyperaminoacidemia associated with reduced kidney glucose output, oxidative stress, enhanced inflammasome activity, and excess lipid accumulation in the kidney. Upon a lipid challenge, they display maladaptive responses with acute hypertriglyceridemia and chronic proinflammatory and profibrotic activation. In aged mice, kidney Gcgr ablation elicits widespread renal deposition of collagen and fibronectin, indicative of fibrosis. Taken together, our findings demonstrate an essential role of the renal GCGR in normal kidney metabolic and homeostatic functions. Importantly, mice deficient for kidney Gcgr recapitulate some of the key pathophysiological features of chronic kidney disease.


Asunto(s)
Receptores de Glucagón , Insuficiencia Renal Crónica , Humanos , Animales , Ratones , Receptores de Glucagón/metabolismo , Regulación hacia Abajo , Ratones Noqueados , Riñón/metabolismo , Homeostasis/fisiología , Lípidos
2.
Diabetes ; 73(2): 197-210, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37935033

RESUMEN

Partial leptin reduction can induce significant weight loss, while weight loss contributes to partial leptin reduction. The cause-and-effect relationship between leptin reduction and weight loss remains to be further elucidated. Here, we show that FGF21 and the glucagon-like peptide 1 receptor (GLP-1R) agonist liraglutide rapidly induced a reduction in leptin. This leptin reduction contributed to the beneficial effects of GLP-1R agonism in metabolic health, as transgenically maintaining leptin levels during treatment partially curtailed the beneficial effects seen with these agonists. Moreover, a higher degree of leptin reduction during treatment, induced by including a leptin neutralizing antibody with either FGF21 or liraglutide, synergistically induced greater weight loss and better glucose tolerance in diet-induced obese mice. Furthermore, upon cessation of either liraglutide or FGF21 treatment, the expected immediate weight regain was observed, associated with a rapid increase in circulating leptin levels. Prevention of this leptin surge with leptin neutralizing antibodies slowed down weight gain and preserved better glucose tolerance. Mechanistically, a significant reduction in leptin induced a higher degree of leptin sensitivity in hypothalamic neurons. Our observations support a model that postulates that a reduction of leptin levels is a necessary prerequisite for substantial weight loss, and partial leptin reduction is a viable strategy to treat obesity and its associated insulin resistance.


Asunto(s)
Leptina , Liraglutida , Animales , Ratones , Leptina/metabolismo , Liraglutida/farmacología , Obesidad , Pérdida de Peso , Glucosa/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo
3.
Sci Transl Med ; 15(723): eade8460, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37992151

RESUMEN

Despite their high degree of effectiveness in the management of psychiatric conditions, exposure to antipsychotic drugs, including olanzapine and risperidone, is frequently associated with substantial weight gain and the development of diabetes. Even before weight gain, a rapid rise in circulating leptin concentrations can be observed in most patients taking antipsychotic drugs. To date, the contribution of this hyperleptinemia to weight gain and metabolic deterioration has not been defined. Here, with an established mouse model that recapitulates antipsychotic drug-induced obesity and insulin resistance, we not only confirm that hyperleptinemia occurs before weight gain but also demonstrate that hyperleptinemia contributes directly to the development of obesity and associated metabolic disorders. By suppressing the rise in leptin through the use of a monoclonal leptin-neutralizing antibody, we effectively prevented weight gain, restored glucose tolerance, and preserved adipose tissue and liver function in antipsychotic drug-treated mice. Mechanistically, suppressing excess leptin resolved local tissue and systemic inflammation typically associated with antipsychotic drug treatment. We conclude that hyperleptinemia is a key contributor to antipsychotic drug-associated weight gain and metabolic deterioration. Leptin suppression may be an effective approach to reducing the undesirable side effects of antipsychotic drugs.


Asunto(s)
Antipsicóticos , Enfermedades Metabólicas , Humanos , Ratones , Animales , Antipsicóticos/efectos adversos , Leptina/metabolismo , Obesidad/metabolismo , Aumento de Peso
4.
Cell Rep ; 40(11): 111362, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36103820

RESUMEN

Obesity is associated with increased cancer incidence and progression. However, the relationship between adiposity and cancer remains poorly understood at the mechanistic level. Here, we report that adipocytes from tumor-invasive mammary fat undergo de-differentiation to fibroblast-like precursor cells during tumor progression and integrate into the tumor microenvironment. Single-cell sequencing reveals that these de-differentiated adipocytes lose their original identities and transform into multiple cell types, including myofibroblast- and macrophage-like cells, with their characteristic features involved in immune response, inflammation, and extracellular matrix remodeling. The de-differentiated cells are metabolically distinct from tumor-associated fibroblasts but exhibit comparable effects on tumor cell proliferation. Inducing de-differentiation by Xbp1s overexpression promotes tumor progression despite lower adiposity. In contrast, promoting lipid-storage capacity in adipocytes through MitoNEET overexpression curbs tumor growth despite greater adiposity. Collectively, the metabolic interplay between tumor cells and adipocytes induces adipocyte mesenchymal transition and contributes to reconfigure the stroma into a more tumor-friendly microenvironment.


Asunto(s)
Neoplasias de la Mama , Neoplasias Mamarias Animales , Adipocitos/metabolismo , Animales , Neoplasias de la Mama/patología , Matriz Extracelular/metabolismo , Femenino , Humanos , Neoplasias Mamarias Animales/patología , Microambiente Tumoral
5.
Acta Pharm Sin B ; 12(7): 3063-3072, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35865093

RESUMEN

Adipose tissue is a promising target for treating obesity and metabolic diseases. However, pharmacological agents usually fail to effectively engage adipocytes due to their extraordinarily large size and insufficient vascularization, especially in obese subjects. We have previously shown that during cold exposure, connexin43 (Cx43) gap junctions are induced and activated to connect neighboring adipocytes to share limited sympathetic neuronal input amongst multiple cells. We reason the same mechanism may be leveraged to improve the efficacy of various pharmacological agents that target adipose tissue. Using an adipose tissue-specific Cx43 overexpression mouse model, we demonstrate effectiveness in connecting adipocytes to augment metabolic efficacy of the ß 3-adrenergic receptor agonist Mirabegron and FGF21. Additionally, combing those molecules with the Cx43 gap junction channel activator danegaptide shows a similar enhanced efficacy. In light of these findings, we propose a model in which connecting adipocytes via Cx43 gap junction channels primes adipose tissue to pharmacological agents designed to engage it. Thus, Cx43 gap junction activators hold great potential for combination with additional agents targeting adipose tissue.

6.
Nat Commun ; 13(1): 1760, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35365663

RESUMEN

The evolutionarily conserved serine/threonine kinase mTORC1 is a central regulator of cell growth and proliferation. mTORC1 is activated on the lysosome surface. However, once mTORC1 is activated, it is unclear whether mTORC1 phosphorylates local lysosomal proteins to regulate specific aspects of lysosomal biology. Through cross-reference analyses of the lysosome proteome with the mTORC1-regulated phosphoproteome, we identify STK11IP as a lysosome-specific substrate of mTORC1. mTORC1 phosphorylates STK11IP at Ser404. Knockout of STK11IP leads to a robust increase of autophagy flux. Dephosphorylation of STK11IP at Ser404 represses the role of STK11IP as an autophagy inhibitor. Mechanistically, STK11IP binds to V-ATPase, and regulates the activity of V-ATPase. Knockout of STK11IP protects mice from fasting or Methionine/Choline-Deficient Diet (MCD)-induced fatty liver. Thus, our study demonstrates that STK11IP phosphorylation represents a mechanism for mTORC1 to regulate lysosomal acidification and autophagy, and points to STK11IP as a promising therapeutic target for the amelioration of diseases with aberrant autophagy signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Complejos Multiproteicos , Serina-Treonina Quinasas TOR , Animales , Concentración de Iones de Hidrógeno , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Noqueados , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
7.
Int J Mol Sci ; 23(7)2022 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-35409189

RESUMEN

Adipocytes from the superficial layer of subcutaneous adipose tissue undergo cyclic de- and re-differentiation, which can significantly influence the development of skin inflammation under different cutaneous conditions. This inflammation can be connected with local loading of the reticular dermis with lipids released due to de-differentiation of adipocytes during the catagen phase of the hair follicle cycle. Alternatively, the inflammation parallels a widespread release of cathelicidin, which typically takes place in the anagen phase (especially in the presence of pathogens). Additionally, trans-differentiation of dermal adipocytes into myofibroblasts, which can occur under some pathological conditions, can be responsible for the development of collateral scarring in acne. Here, we provide an overview of such cellular conversions in the skin and discuss their possible involvement in the pathophysiology of inflammatory skin conditions, such as acne and psoriasis.


Asunto(s)
Acné Vulgar , Enfermedades de la Piel , Adipocitos , Folículo Piloso/fisiología , Humanos , Inflamación , Piel
8.
Bioessays ; 44(1): e2100207, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34766637

RESUMEN

Emerging data connects the aging process in dermal fibroblasts with metabolic reprogramming, provided by enhanced fatty acid oxidation and reduced glycolysis. This switch may be caused by a significant expansion of the dermal white adipose tissue (dWAT) layer in aged, hair-covered skin. Dermal adipocytes cycle through de-differentiation and re-differentiation. As a result, there is a strongly enhanced release of free fatty acids into the extracellular space during the de-differentiation of dermal adipocytes in the catagen phase of the hair follicle cycle. Both caveolin-1 and adiponectin are critical factors influencing these processes. Controlling the expression levels of these two factors also offers the ability to manipulate the metabolic preferences of the different cell types within the microenvironment of the skin, including dermal fibroblasts. Differential expression of adiponectin and caveolin-1 in the various cell types may also be responsible for the cellular metabolic heterogeneity within the cells of the skin.


Asunto(s)
Envejecimiento de la Piel , Adipocitos , Tejido Adiposo Blanco , Fibroblastos , Folículo Piloso , Piel
9.
Nat Commun ; 12(1): 4829, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34376643

RESUMEN

Plasma hyaluronan (HA) increases systemically in type 2 diabetes (T2D) and the HA synthesis inhibitor, 4-Methylumbelliferone, has been proposed to treat the disease. However, HA is also implicated in normal physiology. Therefore, we generated a Hyaluronan Synthase 2 transgenic mouse line, driven by a tet-response element promoter to understand the role of HA in systemic metabolism. To our surprise, adipocyte-specific overproduction of HA leads to smaller adipocytes and protects mice from high-fat-high-sucrose-diet-induced obesity and glucose intolerance. Adipocytes also have more free glycerol that can be released upon beta3 adrenergic stimulation. Improvements in glucose tolerance were not linked to increased plasma HA. Instead, an HA-driven systemic substrate redistribution and adipose tissue-liver crosstalk contributes to the systemic glucose improvements. In summary, we demonstrate an unexpected improvement in glucose metabolism as a consequence of HA overproduction in adipose tissue, which argues against the use of systemic HA synthesis inhibitors to treat obesity and T2D.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Dioxoles/farmacología , Glucosa/metabolismo , Ácido Hialurónico/metabolismo , Lipólisis/efectos de los fármacos , Adipocitos/citología , Tejido Adiposo/citología , Animales , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Femenino , Intolerancia a la Glucosa/metabolismo , Homeostasis , Humanos , Hipoglucemiantes/farmacología , Masculino , Ratones , Ratones Transgénicos , Obesidad/etiología , Obesidad/metabolismo
10.
Cell Metab ; 33(8): 1624-1639.e9, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34174197

RESUMEN

Iron overload is positively associated with diabetes risk. However, the role of iron in adipose tissue remains incompletely understood. Here, we report that transferrin-receptor-1-mediated iron uptake is differentially required for distinct subtypes of adipocytes. Notably, adipocyte-specific transferrin receptor 1 deficiency substantially protects mice from high-fat-diet-induced metabolic disorders. Mechanistically, low cellular iron levels have a positive impact on the health of the white adipose tissue and can restrict lipid absorption from the intestine through modulation of vesicular transport in enterocytes following high-fat diet feeding. Specific reduction of adipocyte iron by AAV-mediated overexpression of the iron exporter Ferroportin1 in adult mice effectively mimics these protective effects. In summary, our studies highlight an important role of adipocyte iron in the maintenance of systemic metabolism through an adipocyte-enterocyte axis, offering an additional level of control over caloric influx into the system after feeding by regulating intestinal lipid absorption.


Asunto(s)
Adipocitos , Tejido Adiposo , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Animales , Dieta Alta en Grasa , Hierro/metabolismo , Lípidos , Ratones , Obesidad/metabolismo
11.
Elife ; 102021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33904399

RESUMEN

Adiponectin is essential for the regulation of tissue substrate utilization and systemic insulin sensitivity. Clinical studies have suggested a positive association of circulating adiponectin with healthspan and lifespan. However, the direct effects of adiponectin on promoting healthspan and lifespan remain unexplored. Here, we are using an adiponectin null mouse and a transgenic adiponectin overexpression model. We directly assessed the effects of circulating adiponectin on the aging process and found that adiponectin null mice display exacerbated age-related glucose and lipid metabolism disorders. Moreover, adiponectin null mice have a significantly shortened lifespan on both chow and high-fat diet. In contrast, a transgenic mouse model with elevated circulating adiponectin levels has a dramatically improved systemic insulin sensitivity, reduced age-related tissue inflammation and fibrosis, and a prolonged healthspan and median lifespan. These results support a role of adiponectin as an essential regulator for healthspan and lifespan.


Asunto(s)
Adiponectina/fisiología , Envejecimiento/metabolismo , Envejecimiento/fisiología , Animales , Femenino , Glucosa/metabolismo , Homeostasis , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos , Longevidad/fisiología , Masculino , Ratones , Ratones Transgénicos
12.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33619103

RESUMEN

We evaluated the potential for a monoclonal antibody antagonist of the glucagon receptor (Ab-4) to maintain glucose homeostasis in type 1 diabetic rodents. We noted durable and sustained improvements in glycemia which persist long after treatment withdrawal. Ab-4 promoted ß-cell survival and enhanced the recovery of insulin+ islet mass with concomitant increases in circulating insulin and C peptide. In PANIC-ATTAC mice, an inducible model of ß-cell apoptosis which allows for robust assessment of ß-cell regeneration following caspase-8-induced diabetes, Ab-4 drove a 6.7-fold increase in ß-cell mass. Lineage tracing suggests that this restoration of functional insulin-producing cells was at least partially driven by α-cell-to-ß-cell conversion. Following hyperglycemic onset in nonobese diabetic (NOD) mice, Ab-4 treatment promoted improvements in C-peptide levels and insulin+ islet mass was dramatically increased. Lastly, diabetic mice receiving human islet xenografts showed stable improvements in glycemic control and increased human insulin secretion.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Diabetes Mellitus Experimental/terapia , Células Secretoras de Glucagón/efectos de los fármacos , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Receptores de Glucagón/antagonistas & inhibidores , Animales , Glucemia/metabolismo , Péptido C/metabolismo , Linaje de la Célula/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/terapia , Expresión Génica , Glucagón/antagonistas & inhibidores , Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/patología , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/fisiología , Trasplante de Islotes Pancreáticos , Ratones , Ratones Endogámicos NOD , Tamaño de los Órganos/efectos de los fármacos , Receptores de Glucagón/genética , Receptores de Glucagón/metabolismo , Resultado del Tratamiento
13.
Exp Dermatol ; 30(1): 102-111, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32866299

RESUMEN

Dermal fibroblasts are an essential population of skin cells. They are not only responsible for synthesis and remodelling of the extracellular matrix of the dermis, but also communicate with other skin cells via autocrine and paracrine interactions. Skin-associated dermal adipocytes reside below the reticular dermis. Strong lipolysis is observed during the regression of dermal adipocytes. However, the nature of the local intercellular crosstalk in which lipids released by dermal adipocytes affecting the metabolism of adjacent skin fibroblasts has not yet been examined. With the use of a series of novel mouse models that allow us to manipulate adipocytes, we demonstrate that dermal adipocytes can modulate the structure of the dermis through regulating extracellular matrix production in dermal fibroblasts. Fatty acids released by dermal adipocytes are involved in this process. Our observations offer new in vivo insights into the role of dermal adipocyte-derived lipids in influencing metabolism of adjacent local cells in the skin through a paracrine effect in the microenvironment of the dermal adipocyte.


Asunto(s)
Adipocitos/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Comunicación Paracrina , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/metabolismo , Animales , Microambiente Celular , Colágeno/genética , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I/genética , Colágeno Tipo III/genética , Células del Cúmulo , Dieta Alta en Grasa , Ácidos Grasos/metabolismo , Femenino , Expresión Génica , Lipólisis , Masculino , Ratones , Piel/citología
14.
Mol Metab ; 39: 101010, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32408016

RESUMEN

OBJECTIVE: Obesity-induced insulin resistance is closely associated with chronic subclinical inflammation in white adipose tissue. However, the mechanistic involvement of adipocyte-derived inflammation under these disease conditions remains unclear. Our aim was to investigate the relative inflammation-related contributions of adipocytes and macrophages to insulin sensitivity. METHODS: RIDα/ß is an adenoviral protein complex that inhibits several inflammatory pathways, including TLR4, TNFα, and IL1ß signaling. We generated novel mouse models with adipocyte-specific and macrophage-specific doxycycline (dox)-inducible RIDα/ß-transgenic mice (RIDad and RIDmac mice, respectively). RESULTS: RIDα/ß induction significantly reduced LPS-stimulated inflammatory markers, such as Tnf, Il1b, and Saa3 in adipose tissues. Surprisingly, RIDad mice had elevated levels of postprandial glucose and insulin and exhibited glucose intolerance and insulin resistance, even under chow-fed conditions. Moreover, the RIDad mice displayed further insulin resistance under obesogenic (high-fat diet, HFD) conditions despite reduced weight gain. In addition, under pre-existing obese and inflamed conditions on an HFD, subsequent induction of RIDα/ß in RIDad mice reduced body weight gain, further exacerbating glucose tolerance, enhancing insulin resistance and fatty liver, and reducing adiponectin levels. This occurred despite effective suppression of the inflammatory pathways (including TNFα and IL1ß). In contrast, RIDmac mice, upon HFD feeding, displayed similar weight gain, comparable adiponectin levels, and insulin sensitivity, suggesting that the inflammatory properties of macrophages did not exert a negative impact on metabolic readouts. RIDα/ß expression and the ensuing suppression of inflammation in adipocytes enhanced adipose tissue fibrosis and reduced vascularization. CONCLUSION: Our novel findings further corroborate our previous observations suggesting that suppressing adipocyte inflammation impairs adipose tissue function and promotes insulin resistance, despite beneficial effects on weight gain.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Inflamación/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Animales , Citocinas/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Inmunohistoquímica , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inflamación/patología , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología
15.
Mol Metab ; 37: 100995, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32289482

RESUMEN

OBJECTIVE: Hyperleptinemia per se is sufficient to promote leptin resistance in the obese state. Leptin sensitivity can be restored by reducing circulating leptin levels within a physiologically healthy range and is a viable antiobesity and antidiabetic strategy. However, a previous study suggests that partial leptin deficiency favors diet-induced obesity and related metabolic disorders in mice, arguing that a lower leptin level may indeed promote diet-induced obesity and its associated metabolic disorders. Here, we aim to elucidate what the impact of partial leptin deficiency is on fat mass and insulin sensitivity. METHODS: We used two different mouse models of partial leptin deficiency: an adipocyte-specific congenital heterozygous leptin knockout mouse line (LepHZ) and the well-established whole body heterozygous leptin knockout mouse (OBHZ). The metabolic studies of OBHZ and LepHZ mice were performed both on normal carbohydrate-rich chow diet and on a high-fat diet (HFD). Male and female mice were included in the study to account for sex-specific differences. Body weight, food intake, glucose tolerance, and insulin tolerance were tested. Histology of adipose tissue and liver tissue allowed insights into adipose tissue inflammation and hepatic triglyceride content. Immunohistochemistry was paired with RT-PCR analysis for expression levels of inflammatory markers. RESULTS: Both OBHZ and LepHZ mice displayed reduced circulating leptin levels on the chow diet and HFD. On chow diet, male OBHZ and LepHZ mice showed elevated fat mass and body weight, while their glucose tolerance and insulin sensitivity remained unchanged. However, the inability in partially leptin-deficient mice to fully induce circulating leptin during the development of diet-induced obesity results in reduced food intake and leaner mice with lower body weight compared to their littermate controls. Importantly, a strong reduction of adipose tissue inflammation is observed along with improvements in insulin sensitivity and enhanced glucose tolerance. Additionally, partial leptin deficiency protects the mice from fatty liver and liver fibrosis. Chronically HFD-fed OBHZ and LepHZ mice remain more sensitive to exogenous leptin injection, as reflected by their reduced food intake upon an acute leptin treatment. CONCLUSION: In response to HFD feeding, the inability to upregulate leptin levels due to partial leptin deficiency protects mice from diet-induced obesity and metabolic dysregulation. Thus, in an obesogenic environment, maintaining lower leptin levels is highly beneficial for both obesity and diabetes management. Chronic leptin reduction represents a viable preventive strategy whose efficacy awaits clinical testing.


Asunto(s)
Leptina/deficiencia , Leptina/metabolismo , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Composición Corporal/fisiología , Peso Corporal/fisiología , Dieta Alta en Grasa , Hígado Graso/fisiopatología , Femenino , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Hígado/patología , Masculino , Enfermedades Metabólicas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/prevención & control
16.
Cell Metab ; 30(4): 706-719.e6, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31495688

RESUMEN

The physiological role of leptin is thought to be a driving force to reduce food intake and increase energy expenditure. However, leptin therapies in the clinic have failed to effectively treat obesity, predominantly due to a phenomenon referred to as leptin resistance. The mechanisms linking obesity and the associated leptin resistance remain largely unclear. With various mouse models and a leptin neutralizing antibody, we demonstrated that hyperleptinemia is a driving force for metabolic disorders. A partial reduction of plasma leptin levels in the context of obesity restores hypothalamic leptin sensitivity and effectively reduces weight gain and enhances insulin sensitivity. These results highlight that a partial reduction in plasma leptin levels leads to improved leptin sensitivity, while pointing to a new avenue for therapeutic interventions in the treatment of obesity and its associated comorbidities.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Resistencia a la Insulina , Insulina/metabolismo , Leptina/antagonistas & inhibidores , Obesidad/terapia , Pérdida de Peso/efectos de los fármacos , Programas de Reducción de Peso/métodos , Animales , Anticuerpos Neutralizantes/uso terapéutico , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Leptina/sangre , Ratones , Ratones Endogámicos , Obesidad/metabolismo
17.
J Clin Invest ; 129(12): 5327-5342, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31503545

RESUMEN

Dermal adipose tissue (also known as dermal white adipose tissue and herein referred to as dWAT) has been the focus of much discussion in recent years. However, dWAT remains poorly characterized. The fate of the mature dermal adipocytes and the origin of the rapidly reappearing dermal adipocytes at different stages remain unclear. Here, we isolated dermal adipocytes and characterized dermal fat at the cellular and molecular level. Together with dWAT's dynamic responses to external stimuli, we established that dermal adipocytes are a distinct class of white adipocytes with high plasticity. By combining pulse-chase lineage tracing and single-cell RNA sequencing, we observed that mature dermal adipocytes undergo dedifferentiation and redifferentiation under physiological and pathophysiological conditions. Upon various challenges, the dedifferentiated cells proliferate and redifferentiate into adipocytes. In addition, manipulation of dWAT highlighted an important role for mature dermal adipocytes for hair cycling and wound healing. Altogether, these observations unravel a surprising plasticity of dermal adipocytes and provide an explanation for the dynamic changes in dWAT mass that occur under physiological and pathophysiological conditions, and highlight the important contributions of dWAT toward maintaining skin homeostasis.


Asunto(s)
Adipocitos Blancos/citología , Desdiferenciación Celular/fisiología , Plasticidad de la Célula/fisiología , Piel/citología , Adipocitos Blancos/fisiología , Animales , Diferenciación Celular , Separación Celular , Perfilación de la Expresión Génica , Folículo Piloso/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/citología , Cicatrización de Heridas
18.
Ageing Res Rev ; 55: 100959, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31493519

RESUMEN

Caveolin-1 (Cav-1) appears to be both a pathophysiological contributor and a target in different inflammatory and hyperproliferative skin conditions as well as in skin aging. Skin fibroblasts demonstrate an up-regulation of Cav-1 expression both in chronological and UV-induced aging, and such an up-regulation was observed both in vitro and in vivo. Typical alterations in aging skin involve a reduction of the dermis thickness, a significant expansion of the dermal white adipose tissue as well as modifications of the content and distribution of hyaluronan, impairment of autophagic flux, a reduction of collagen expression and an increase in tissue inflammation. All of these phenomena can be connected with changes in Cav-1 expression in the aging skin. Modified expression of Cav-1 can also significantly influence the mechanical properties of individual skin layers, thus changing the total mechanical stability of the layered composite skin/WAT, leading to typical structural modifications of the skin surface in the aging skin. Selective reduction of Cav-1 expression has the potential to exert anti-aging effects on the skin.


Asunto(s)
Caveolina 1/fisiología , Envejecimiento de la Piel , Envejecimiento , Animales , Fibroblastos/metabolismo , Fibroblastos/fisiología , Humanos , Piel/metabolismo
19.
Nutr Metab (Lond) ; 16: 41, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31303888

RESUMEN

BACKGROUND: Apolipoprotein A-IV (ApoA-IV) exists in relatively high levels in the circulation systems of animals, but its roles are not fully elucidated. It is known that the Apoa4 gene resides in the cluster Apoa1/Apoc3/Apoa4. Because of a short intergenic sequence between Apoc3 and Apoa4, a previous ApoA-IV knockout mouse model by gene targeting had an accompanying deficiency in ApoC-III expression, which limited its application in investigating the precise roles of ApoA-IV. To solve this problem, we created a specific knockout of ApoA-IV in Sprague-Dawlay rats by TALEN approach. METHODS: Age-matched knockout rats and their wild-type littermate controls maintained on a standard rodent diet were studied and blood metabolic parameters were measured. Glucose, insulin, olive oil, and intralipid tolerance tests were performed to study the glucose and lipid metabolism of rats. Quantitative real-time PCR and RNA-seq analysis in liver and inguinal white adipose tissue (iWAT) of rats at three ages (18 weeks, 45 weeks and 90 weeks) were performed to identify the genes altered by ApoA-IV knockout. RESULTS: ApoA-IV knockout rats were apparently normal and fertile, but exhibited improved glucose clearance when challenged with glucose tolerance test. In addition, fasting-induced hepatic steatosis was more pronounced in ApoA-IV knockout rats. Further analysis identified that a set of hepatic genes involved in glycolysis, gluconeogenesis and de novo lipogenesis were altered in the absence of ApoA-IV, which induced enhanced glycolysis, attenuated gluconeogenesis and elevated de novo lipogenesis. And the RNA-seq results also confirmed that almost all the genes mentioned in the phenotyping section were highly consistent throughout the three studied ages. CONCLUSIONS: ApoA-IV functions in an age-independent manner in the modulation of glucose and lipid metabolism of rats, and may serve as a potential linker between hepatic glucose and lipid metabolism.

20.
PLoS Genet ; 15(3): e1008021, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30830908

RESUMEN

[This corrects the article DOI: 10.1371/journal.pgen.1005485.].

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...