Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.442
Filtrar
1.
Biotechnol J ; 19(5): e2300676, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38730523

RESUMEN

Genetic diseases can be caused by monogenic diseases, which result from a single gene mutation in the DNA sequence. Many innovative approaches have been developed to cure monogenic genetic diseases, namely by genome editing. A specific type of genomic editing, prime editing, has the potential advantage to edit the human genome without requiring double-strand breaks or donor DNA templates for editing. Additionally, prime editing does not require a precisely positioned protospacer adjacent motif (PAM) sequence, which offers flexible target and more precise genomic editing. Here we detail a novel construction of a prime editing extended guide RNA (pegRNA) to target mutated leptin receptors in B6.BKS(D)-Leprdb/J mice (db/db mice). The pegRNA was then injected into the flexor digitorum brevis (FDB) muscle of db/db mice to demonstrate in vivo efficacy, which resulted in pegRNA mediated base transversion at endogenous base transversion. Genomic DNA sequencing confirmed that prime editing could correct the mutation of leptin receptor gene in db/db mice. Furthermore, prime editing treated skeletal muscle exhibited enhanced leptin receptor signals. Thus, the current study showed in vivo efficacy of prime editing to correct mutant protein and rescue the physiology associated with functional protein.


Asunto(s)
Edición Génica , Receptores de Leptina , Animales , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Edición Génica/métodos , Ratones , Músculo Esquelético/metabolismo , ARN Guía de Sistemas CRISPR-Cas/genética , Mutación , Sistemas CRISPR-Cas/genética , Ratones Endogámicos C57BL
2.
Arq Gastroenterol ; 61: e24016, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38775586

RESUMEN

BACKGROUND: Colorectal carcinoma (CRC) is one of the common carcinomas with a rising incidence of metastasis due to its advanced stage of presentation. The existing biomarkers such as CEA (Carcinoembryonic antigen) etc., for prognosis, have low sensitivity and specificity. Hence a need for a newer definitive biomarker. Obesity is the leading cause of CRC. Leptin and adiponectin secreted by adipose tissue have been studied as potential biomarkers in the field of CRC. The present study helps to understand the association of leptin and adiponectin receptors with clinicopathological parameters. OBJECTIVE: To correlate the various clinicopathological parameters with the tissue expression of leptin and adiponectin receptors in CRC. METHODS: It is a cross-sectional prospective study conducted at a tertiary care hospital. Formalin fixed paraffin blocks of all radical resection CRC cases were collected and immunohistochemistry (IHC)was carried out on tumor tissue for leptin and adiponectin receptor. Tumor characteristics and clinical parameters were collected from the hospital medical records. Pearson's correlation coefficient test was used. RESULTS: Immunohistochemistry was performed on 60 cases of CRC. Significant positive correlation of leptin was observed with size, lymph node metastasis, advanced stage, and grade of tumor (P<0.05). A significant correlation between adiponectin receptor and CRC was observed concerning age, stage, lymph node metastasis, distant metastasis and grade of tumor. CONCLUSION: Positive expression of leptin and negative expression of adiponectin receptors in CRC helps to predict the risk of metastasis.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Colorrectales , Inmunohistoquímica , Leptina , Estadificación de Neoplasias , Receptores de Adiponectina , Humanos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Estudios Transversales , Estudios Prospectivos , Masculino , Femenino , Persona de Mediana Edad , Leptina/metabolismo , Leptina/análisis , Receptores de Adiponectina/análisis , Receptores de Adiponectina/metabolismo , Anciano , Biomarcadores de Tumor/metabolismo , Adulto , Receptores de Leptina/metabolismo , Receptores de Leptina/análisis , Clasificación del Tumor , Metástasis Linfática
3.
Life Sci ; 346: 122649, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38626868

RESUMEN

AIMS: Leptin irresponsiveness, which is often associated with obesity, can have significant impacts on the hypothalamic proteome of individuals, including those who are lean. While mounting evidence on leptin irresponsiveness has focused on obese individuals, understanding the early molecular and proteomic changes associated with deficient hypothalamic leptin signaling in lean individuals is essential for early intervention and prevention of metabolic disorders. Leptin receptor antagonists block the binding of leptin to its receptors, potentially reducing its effects and used in cases where excessive leptin activity might be harmful. MATERIALS AND METHODS: In this work, we blocked the central actions of leptin in lean male adult Wistar rat by chronically administering intracerebroventricularly the superactive leptin receptor antagonist (SLA) (D23L/L39A/D40A/F41A) and investigated its impact on the hypothalamic proteome using label-free sequential window acquisition of all theoretical fragment ion spectra mass spectrometry (SWATH-MS) for quantitative proteomics. KEY FINDINGS: Our results show an accumulation of proteins involved in mRNA processing, mRNA stability, and translation in the hypothalamus of SLA-treated rats. Conversely, hypothalamic leptin signaling deficiency reduces the representation of proteins implicated in energy metabolism, neural circuitry, and neurotransmitter release. SIGNIFICANCE: The alterations in the adult rat hypothalamic proteome contribute to dysregulate appetite, metabolism, and energy balance, which are key factors in the development and progression of obesity and related metabolic disorders. Additionally, using bioinformatic analysis, we identified a series of transcription factors that are potentially involved in the upstream regulatory mechanisms responsible for the observed signature.


Asunto(s)
Hipotálamo , Leptina , Proteoma , Proteómica , Ratas Wistar , Receptores de Leptina , Transducción de Señal , Animales , Masculino , Leptina/metabolismo , Receptores de Leptina/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/deficiencia , Hipotálamo/metabolismo , Hipotálamo/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos , Proteómica/métodos , Proteoma/metabolismo , Obesidad/metabolismo , Metabolismo Energético/efectos de los fármacos
4.
Nutrients ; 16(8)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38674862

RESUMEN

Leptin is an appetite-regulating adipokine that is reduced in patients with anorexia nervosa (AN), a psychiatric disorder characterized by self-imposed starvation, and has been linked to hyperactivity, a hallmark of AN. However, it remains unknown how leptin receptor (LepR) and its JAK2-STAT3 downstream pathway in extrahypothalamic brain areas, such as the dorsal (dHip) and ventral (vHip) hippocampus, crucial for spatial memory and emotion regulation, may contribute to the maintenance of AN behaviors. Taking advantage of the activity-based anorexia (ABA) model (i.e., the combination of food restriction and physical activity), we observed reduced leptin plasma levels in adolescent female ABA rats at the acute phase of the disorder [post-natal day (PND) 42], while the levels increased over control levels following a 7-day recovery period (PND49). The analysis of the intracellular leptin pathway revealed that ABA rats showed an overall decrease of the LepR/JAK2/STAT3 signaling in dHip at both time points, while in vHip we observed a transition from hypo- (PND42) to hyperactivation (PND49) of the pathway. These changes might add knowledge on starvation-induced fluctuations in leptin levels and in hippocampal leptin signaling as initial drivers of the transition from adaptative mechanisms to starvation toward the maintenance of aberrant behaviors typical of AN patients, such as perpetuating restraint over eating.


Asunto(s)
Anorexia , Hipocampo , Janus Quinasa 2 , Leptina , Receptores de Leptina , Factor de Transcripción STAT3 , Transducción de Señal , Animales , Femenino , Janus Quinasa 2/metabolismo , Factor de Transcripción STAT3/metabolismo , Hipocampo/metabolismo , Leptina/sangre , Anorexia/etiología , Anorexia/metabolismo , Ratas , Receptores de Leptina/metabolismo , Anorexia Nerviosa/metabolismo , Anorexia Nerviosa/sangre , Modelos Animales de Enfermedad , Adaptación Fisiológica
5.
Cell Metab ; 36(4): 857-876.e10, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38569472

RESUMEN

Leptin resistance during excess weight gain significantly contributes to the recidivism of obesity to leptin-based pharmacological therapies. The mechanisms underlying the inhibition of leptin receptor (LepR) signaling during obesity are still elusive. Here, we report that histone deacetylase 6 (HDAC6) interacts with LepR, reducing the latter's activity, and that pharmacological inhibition of HDAC6 activity disrupts this interaction and augments leptin signaling. Treatment of diet-induced obese mice with blood-brain barrier (BBB)-permeable HDAC6 inhibitors profoundly reduces food intake and leads to potent weight loss without affecting the muscle mass. Genetic depletion of Hdac6 in Agouti-related protein (AgRP)-expressing neurons or administration with BBB-impermeable HDAC6 inhibitors results in a lack of such anti-obesity effect. Together, these findings represent the first report describing a mechanistically validated and pharmaceutically tractable therapeutic approach to directly increase LepR activity as well as identifying centrally but not peripherally acting HDAC6 inhibitors as potent leptin sensitizers and anti-obesity agents.


Asunto(s)
Leptina , Obesidad , Animales , Ratones , Histona Desacetilasa 6 , Leptina/metabolismo , Obesidad/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Aumento de Peso , Pérdida de Peso
6.
Biomolecules ; 14(4)2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38672473

RESUMEN

There is a high demand for agonist biomolecules such as cytokine surrogates in both biological and medicinal research fields. These are typically sourced through natural ligand engineering or affinity-based screening, followed by individual functional validation. However, efficient screening methods for identifying rare hits within immense libraries are very limited. In this research article, we introduce a phenotypic screening method utilizing biological receptor activation-dependent cell survival (BRADS). This method offers a high-throughput, low-background, and cost-effective approach that can be implemented in virtually any biochemical laboratory setting. As a proof-of-concept, we successfully identified a surrogate for human leptin following a two-week cell culture process, without the need for specialized high-throughput equipment or reagents. This surrogate effectively emulates the activity of native human leptin in cell validation assays. Our findings not only underscore the effectiveness of BRADS but also suggest its potential applicability to a broad range of biological receptors, including Notch and GPCRs.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Leptina , Leptina/metabolismo , Humanos , Ensayos Analíticos de Alto Rendimiento/métodos , Supervivencia Celular/efectos de los fármacos , Fenotipo , Células HEK293 , Ligandos , Receptores de Leptina/agonistas , Receptores de Leptina/metabolismo
7.
Int Immunopharmacol ; 132: 111990, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38574702

RESUMEN

Hydrogen sulfide (H2S), recognized as the third gasotransmitter, plays a pivotal role in the pathophysiological processes of various diseases. Cystathionine γ-lyase (CSE) is the main enzyme for H2S production in the skin. However, effects and mechanisms of H2S in diabetic skin wound healing remain unclear. Our findings revealed a decrease in plasma H2S content in diabetic patients with skin wounds. CSE knockout (KO) diabetic mice resulted in delayed wound healing, reduced blood perfusion, and CD31 expression around the wounds. It also led to increased infiltration of inflammatory cells and M1-type macrophages, decreased collagen levels, α-smooth muscle actin (α-SMA), and proliferating cell nuclear antigen (PCNA) expression. Additionally, there were enhanced expressions of necroptosis related proteins, including receptor interacting protein kinase 1 (RIPK1), RIPK3 and mixed lineage kinase domain like protein (MLKL). In comparison, sodium hydrosulfide (NaHS), H2S donor, accelerated skin wound healing in leptin receptor deficiency (db/db) mice. This acceleration was accompanied by increased blood perfusion and CD31 expression, reduced infiltration of inflammatory cells and M1-type macrophages, elevated collagen levels, α-SMA, and PCNA expressions, and decreased necroptosis-related protein expressions together with nuclear factor-κB (NF-κB) p65 phosphorylation. In conclusion, H2S regulates macrophage polarization and necroptosis, contributing to the acceleration of diabetic skin wound healing. These findings offer a novel strategy for the treatment of diabetic skin wounds.


Asunto(s)
Cistationina gamma-Liasa , Diabetes Mellitus Experimental , Sulfuro de Hidrógeno , Macrófagos , Ratones Endogámicos C57BL , Ratones Noqueados , Necroptosis , Piel , Sulfuros , Cicatrización de Heridas , Animales , Sulfuro de Hidrógeno/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Piel/patología , Piel/metabolismo , Piel/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Diabetes Mellitus Experimental/metabolismo , Cistationina gamma-Liasa/metabolismo , Cistationina gamma-Liasa/genética , Masculino , Ratones , Humanos , Necroptosis/efectos de los fármacos , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
8.
Mol Metab ; 83: 101915, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38492844

RESUMEN

OBJECTIVE: The glucose-dependent insulinotropic polypeptide (GIP) decreases body weight via central GIP receptor (GIPR) signaling, but the underlying mechanisms remain largely unknown. Here, we assessed whether GIP regulates body weight and glucose control via GIPR signaling in cells that express the leptin receptor (Lepr). METHODS: Hypothalamic, hindbrain, and pancreatic co-expression of Gipr and Lepr was assessed using single cell RNAseq analysis. Mice with deletion of Gipr in Lepr cells were generated and metabolically characterized for alterations in diet-induced obesity (DIO), glucose control and leptin sensitivity. Long-acting single- and dual-agonists at GIPR and GLP-1R were further used to assess drug effects on energy and glucose metabolism in DIO wildtype (WT) and Lepr-Gipr knock-out (KO) mice. RESULTS: Gipr and Lepr show strong co-expression in the pancreas, but not in the hypothalamus and hindbrain. DIO Lepr-Gipr KO mice are indistinguishable from WT controls related to body weight, food intake and diet-induced leptin resistance. Acyl-GIP and the GIPR:GLP-1R co-agonist MAR709 remain fully efficacious to decrease body weight and food intake in DIO Lepr-Gipr KO mice. Consistent with the demonstration that Gipr and Lepr highly co-localize in the endocrine pancreas, including the ß-cells, we find the superior glycemic effect of GIPR:GLP-1R co-agonism over single GLP-1R agonism to vanish in Lepr-Gipr KO mice. CONCLUSIONS: GIPR signaling in cells/neurons that express the leptin receptor is not implicated in the control of body weight or food intake, but is of crucial importance for the superior glycemic effects of GIPR:GLP-1R co-agonism relative to single GLP-1R agonism.


Asunto(s)
Peso Corporal , Ingestión de Alimentos , Polipéptido Inhibidor Gástrico , Ratones Noqueados , Obesidad , Receptores de la Hormona Gastrointestinal , Receptores de Leptina , Animales , Masculino , Ratones , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/genética , Glucosa/metabolismo , Leptina/metabolismo , Ratones Endogámicos C57BL , Obesidad/metabolismo , Receptores de la Hormona Gastrointestinal/metabolismo , Receptores de la Hormona Gastrointestinal/genética , Receptores de Leptina/metabolismo , Receptores de Leptina/genética , Transducción de Señal
9.
Neuropeptides ; 105: 102418, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38442503

RESUMEN

The aim of this study is to verify the impact of Leptin in blood pressure (BP) regulation and Leptin-resistance in metabolic/neurogenic hypertension through baroreflex afferents and dysregulation. Artery BP/heart rate (HR) were measured while nodose (NG) microinjection of Leptin, membrane depolarization/inward current were obtained by whole-cell patch from NG neurons isolated from adult female rats. Baroreflex sensitivity (BRS) tested with PE/SNP, distribution/expression of Leptin/receptors in the NG/nucleus tractus solitary (NTS) examined using immumostaining and qRT-PCR, and serum concentrations of Leptin/NE measured by ELISA were observed in control and high fructose-drinking induced hypertension (HTN-HFD) rats. The results showed that BP was significantly/dose-dependently reduced by Leptin NG microinjection likely through direct excitation of female-specific subpopulation of Ah-type neurons showing a potent membrane depolarization/inward currents. Sex-specific distribution/expression of OB-Ra/OB-Rb in the NG were detected with estrogen-dependent manner, similar observations were also confirmed in the NTS. As expected, BRS was dramatically decreased in the presence of PE/SNP in both male and female rats except for the female with PE at given concentrations. Additionally, serum concentration of Leptin was elevated in HFD-HTN model rats of either sex with more obvious in females. Under hypertensive condition, the mean fluorescent density of OB-R and mRNA expression for OB-Ra/OB-Rb in the NG/NTS were significantly down-regulated. These results have demonstrated that Leptin play a role in dominant parasympathetic drive via baroreflex afferent activation to buffer Leptin-mediated sympathetic activation systemically and Leptin-resistance is an innegligible mechanism for metabolic/neurogenic hypertension through baroreflex afferent dysregulation.


Asunto(s)
Barorreflejo , Presión Sanguínea , Hipertensión , Leptina , Ratas Sprague-Dawley , Animales , Leptina/farmacología , Leptina/metabolismo , Leptina/sangre , Barorreflejo/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Hipertensión/metabolismo , Hipertensión/fisiopatología , Femenino , Masculino , Ratas , Ganglio Nudoso/metabolismo , Ganglio Nudoso/efectos de los fármacos , Núcleo Solitario/metabolismo , Núcleo Solitario/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Receptores de Leptina/metabolismo , Neuronas/metabolismo , Neuronas/efectos de los fármacos
10.
J Neurophysiol ; 131(4): 689-708, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38416718

RESUMEN

Metabolic syndrome (MetS) is associated with development of tauopathies that contribute to cognitive decline. Without functional leptin receptors, male obese Zucker rats (OZRs) develop MetS, and they have increased phosphorylated tau (ptau) with impaired cognitive function. In addition to regulating energy balance, leptin enhances activation of the hippocampus, which is essential for spatial learning and memory. Whether spatial learning and memory are always impaired in OZRs or develop with MetS is unknown. We hypothesized that male OZRs develop MetS traits that promote regional increases in ptau and functional deficits associated with those brain regions. In the medulla and cortex, tau-pSer199,202 and tau-pSer396 were comparable in juvenile (7-8 wk old) lean Zucker rats (LZRs) and OZRs but increased in 18- to 19-wk-old OZRs. Elevated tau-pSer396 was concentrated in the dorsal vagal complex of the medulla, and by this age OZRs had hypertension with increased arterial pressure variability. In the hippocampus, tau-pSer199,202 and tau-pSer396 were still comparable in 18- to 19-wk-old OZRs and LZRs but elevated in 28- to 29-wk-old OZRs, with emergence of deficits in Morris water maze performance. Comparable escape latencies observed during acquisition in 18- to 19-wk-old OZRs and LZRs were increased in 28- to 29-wk-old OZRs, with greater use of nonspatial search strategies. Increased ptau developed with changes in the insulin/phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway in the hippocampus and cortex but not medulla, suggesting different underlying mechanisms. These data demonstrate that leptin is not required for spatial learning and memory in male OZRs. Furthermore, early development of MetS-associated autonomic dysfunction by the medulla may be predictive of later hippocampal dysfunction and cognitive impairment.NEW & NOTEWORTHY Male obese Zucker rats (OZRs) lack functional leptin receptors and develop metabolic syndrome (MetS). At 16-19 wk, OZRs are insulin resistant, with increased ptau in dorsal medulla and impaired autonomic regulation of AP. At 28-29 wk OZRs develop increased ptau in hippocampus with deficits in spatial learning and memory. Juvenile OZRs lack elevated ptau and these deficits, demonstrating that leptin is not essential for normal function. Elevated ptau and deficits emerge before the onset of diabetes in insulin-resistant OZRs.


Asunto(s)
Hipertensión , Síndrome Metabólico , Animales , Ratas , Masculino , Síndrome Metabólico/complicaciones , Leptina/metabolismo , Ratas Zucker , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Leptina/metabolismo , Obesidad , Insulina , Prosencéfalo , Modelos Animales de Enfermedad , Hipocampo/metabolismo
11.
J Neuroendocrinol ; 36(2): e13366, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38279680

RESUMEN

The arcuate nucleus is a crucial hypothalamic brain region involved in regulating body weight homeostasis. Neurons within the arcuate nucleus respond to peripheral metabolic signals, such as leptin, and relay these signals via neuronal projections to brain regions both within and outside the hypothalamus, ultimately causing changes in an animal's behaviour and physiology. There is a substantial amount of evidence to indicate that leptin is intimately involved with the postnatal development of arcuate nucleus melanocortin circuitry. Further, it is clear that leptin signalling directly in the arcuate nucleus is required for circuitry development. However, as leptin receptor long isoform (Leprb) mRNA is expressed in multiple nuclei within the developing hypothalamus, including the postsynaptic target regions of arcuate melanocortin projections, this raises the possibility that leptin also signals in these nuclei to promote circuitry development. Here, we used RT-qPCR and RNAscope® to reveal the spatio-temporal pattern of Leprb mRNA in the early postnatal mouse hypothalamus. We found that Leprb mRNA expression increased significantly in the arcuate nucleus, ventromedial nucleus and paraventricular nucleus of the hypothalamus from P8, in concert with the leptin surge. In the dorsomedial nucleus of the hypothalamus, increases in Leprb mRNA were slightly later, increasing significantly from P12. Using duplex RNAscope®, we found Leprb co-expressed with Sim1, Pou3f2, Mc4r and Bdnf in the paraventricular nucleus at P8. Together, these data suggest that leptin may signal in a subset of neurons postsynaptic to arcuate melanocortin neurons, as well as within the arcuate nucleus itself, to promote the formation of arcuate melanocortin circuitry during the early postnatal period.


Asunto(s)
Leptina , Receptores de Leptina , Animales , Ratones , Leptina/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Melanocortinas/metabolismo , ARN Mensajero/metabolismo
12.
Immunity ; 57(1): 141-152.e5, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38091996

RESUMEN

Adipose tissues (ATs) are innervated by sympathetic nerves, which drive reduction of fat mass via lipolysis and thermogenesis. Here, we report a population of immunomodulatory leptin receptor-positive (LepR+) sympathetic perineurial barrier cells (SPCs) present in mice and humans, which uniquely co-express Lepr and interleukin-33 (Il33) and ensheath AT sympathetic axon bundles. Brown ATs (BATs) of mice lacking IL-33 in SPCs (SPCΔIl33) had fewer regulatory T (Treg) cells and eosinophils, resulting in increased BAT inflammation. SPCΔIl33 mice were more susceptible to diet-induced obesity, independently of food intake. Furthermore, SPCΔIl33 mice had impaired adaptive thermogenesis and were unresponsive to leptin-induced rescue of metabolic adaptation. We therefore identify LepR+ SPCs as a source of IL-33, which orchestrate an anti-inflammatory BAT environment, preserving sympathetic-mediated thermogenesis and body weight homeostasis. LepR+IL-33+ SPCs provide a cellular link between leptin and immune regulation of body weight, unifying neuroendocrinology and immunometabolism as previously disconnected fields of obesity research.


Asunto(s)
Tejido Adiposo Pardo , Leptina , Animales , Humanos , Ratones , Tejido Adiposo Pardo/inervación , Tejido Adiposo Pardo/metabolismo , Peso Corporal , Metabolismo Energético/fisiología , Interleucina-33/genética , Interleucina-33/metabolismo , Obesidad/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Termogénesis/fisiología
13.
J Cell Physiol ; 239(2): e31117, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37683049

RESUMEN

Leptin is an adipocyte-derived hormone that modulates food intake, energy balance, neuroendocrine status, thermogenesis, and cognition. Whereas a high density of leptin receptors has been detected in the basolateral amygdala (BLA) neurons, the physiological functions of leptin in the BLA have not been determined yet. We found that application of leptin excited BLA principal neurons by activation of the long form leptin receptor, LepRb. The LepRb-elicited excitation of BLA neurons was mediated by depression of the G protein-activated inwardly rectifying potassium (GIRK) channels. Janus Kinase 2 (JAK2) and phosphoinositide 3-kinase (PI3K) were required for leptin-induced excitation of BLA neurons and depression of GIRK channels. Microinjection of leptin into the BLA reduced food intake via activation of LepRb, JAK2, and PI3K. Our results may provide a cellular and molecular mechanism to explain the physiological roles of leptin in vivo.


Asunto(s)
Complejo Nuclear Basolateral , Fosfatidilinositol 3-Quinasas , Complejo Nuclear Basolateral/metabolismo , Ingestión de Alimentos , Janus Quinasa 2 , Leptina/farmacología , Leptina/metabolismo , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasa , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Masculino , Femenino , Animales , Ratas , Ratas Sprague-Dawley , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo
14.
Am J Physiol Regul Integr Comp Physiol ; 326(5): R383-R400, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38105761

RESUMEN

The hormone leptin reduces food intake through actions in the peripheral and central nervous systems, including in the hindbrain nucleus of the solitary tract (NTS). The NTS receives viscerosensory information via vagal afferents, including information from the gastrointestinal tract, which is then relayed to other central nervous system (CNS) sites critical for control of food intake. Leptin receptors (lepRs) are expressed by a subpopulation of NTS neurons, and knockdown of these receptors increases both food intake and body weight. Recently, we demonstrated that leptin increases vagal activation of lepR-expressing neurons via increased NMDA receptor (NMDAR) currents, thereby potentiating vagally evoked firing. Furthermore, chemogenetic activation of these neurons was recently shown to inhibit food intake. However, the vagal inputs these neurons receive had not been characterized. Here we performed whole cell recordings in brain slices taken from lepRCre × floxedTdTomato mice and found that lepR neurons of the NTS are directly activated by monosynaptic inputs from C-type afferents sensitive to the transient receptor potential vanilloid type 1 (TRPV1) agonist capsaicin. CCK administered onto NTS slices stimulated spontaneous glutamate release onto lepR neurons and induced action potential firing, an effect mediated by CCKR1. Interestingly, NMDAR activation contributed to the current carried by spontaneous excitatory postsynaptic currents (EPSCs) and enhanced CCK-induced firing. Peripheral CCK also increased c-fos expression in these neurons, suggesting they are activated by CCK-sensitive vagal afferents in vivo. Our results indicate that the majority of NTS lepR neurons receive direct inputs from CCK-sensitive C vagal-type afferents, with both peripheral and central CCK capable of activating these neurons and NMDARs able to potentiate these effects.


Asunto(s)
Receptores de N-Metil-D-Aspartato , Núcleo Solitario , Animales , Ratones , Leptina/metabolismo , Fibras Nerviosas Amielínicas/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Núcleo Solitario/metabolismo , Nervio Vago/fisiología
15.
Proc Natl Acad Sci U S A ; 121(1): e2310685120, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38147550

RESUMEN

Leptin protein was thought to be unique to leptin receptor (LepR), but the phenotypes of mice with mutation in LepR [db/db (diabetes)] and leptin [ob/ob (obese)] are not identical, and the cause remains unclear. Here, we show that db/db, but not ob/ob, mice had defect in tenotomy-induced heterotopic ossification (HO), implicating alternative ligand(s) for LepR might be involved. Ligand screening revealed that ANGPTL4 (angiopoietin-like protein 4), a stress and fasting-induced factor, was elicited from brown adipose tissue after tenotomy, bound to LepR on PRRX1+ mesenchymal cells at the HO site, thus promotes chondrogenesis and HO development. Disruption of LepR in PRRX1+ cells, or lineage ablation of LepR+ cells, or deletion of ANGPTL4 impeded chondrogenesis and HO in mice. Together, these findings identify ANGPTL4 as a ligand for LepR to regulate the formation of acquired HO.


Asunto(s)
Leptina , Osificación Heterotópica , Animales , Ratones , Leptina/genética , Ligandos , Ratones Endogámicos C57BL , Osteogénesis , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
16.
Cell Rep ; 42(12): 113512, 2023 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-38039129

RESUMEN

Mismatch between CO2 production (Vco2) and respiration underlies the pathogenesis of obesity hypoventilation. Leptin-mediated CNS pathways stimulate both metabolism and breathing, but interactions between these functions remain elusive. We hypothesized that LEPRb+ neurons of the dorsomedial hypothalamus (DMH) regulate metabolism and breathing in obesity. In diet-induced obese LeprbCre mice, chemogenetic activation of LEPRb+ DMH neurons increases minute ventilation (Ve) during sleep, the hypercapnic ventilatory response, Vco2, and Ve/Vco2, indicating that breathing is stimulated out of proportion to metabolism. The effects of chemogenetic activation are abolished by a serotonin blocker. Optogenetic stimulation of the LEPRb+ DMH neurons evokes excitatory postsynaptic currents in downstream serotonergic neurons of the dorsal raphe (DR). Administration of retrograde AAV harboring Cre-dependent caspase to the DR deletes LEPRb+ DMH neurons and abolishes metabolic and respiratory responses to leptin. These findings indicate that LEPRb+ DMH neurons match breathing to metabolism through serotonergic pathways to prevent obesity-induced hypoventilation.


Asunto(s)
Hipoventilación , Leptina , Ratones , Animales , Leptina/metabolismo , Hipoventilación/metabolismo , Obesidad/metabolismo , Respiración , Hipotálamo/metabolismo , Receptores de Leptina/metabolismo
17.
Nat Cell Biol ; 25(12): 1746-1757, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38012403

RESUMEN

The bone marrow contains peripheral nerves that promote haematopoietic regeneration after irradiation or chemotherapy (myeloablation), but little is known about how this is regulated. Here we found that nerve growth factor (NGF) produced by leptin receptor-expressing (LepR+) stromal cells is required to maintain nerve fibres in adult bone marrow. In nerveless bone marrow, steady-state haematopoiesis was normal but haematopoietic and vascular regeneration were impaired after myeloablation. LepR+ cells, and the adipocytes they gave rise to, increased NGF production after myeloablation, promoting nerve sprouting in the bone marrow and haematopoietic and vascular regeneration. Nerves promoted regeneration by activating ß2 and ß3 adrenergic receptor signalling in LepR+ cells, and potentially in adipocytes, increasing their production of multiple haematopoietic and vascular regeneration growth factors. Peripheral nerves and LepR+ cells thus promote bone marrow regeneration through a reciprocal relationship in which LepR+ cells sustain nerves by synthesizing NGF and nerves increase regeneration by promoting the production of growth factors by LepR+ cells.


Asunto(s)
Médula Ósea , Receptores de Leptina , Médula Ósea/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Células de la Médula Ósea/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Células Madre Hematopoyéticas/metabolismo , Regeneración Nerviosa
18.
Sci Transl Med ; 15(723): eadd4897, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37992152

RESUMEN

Deficiency in the adipose-derived hormone leptin or leptin receptor signaling causes class 3 obesity in individuals with genetic loss-of-function mutations in leptin or its receptor LEPR and metabolic and liver disease in individuals with hypoleptinemia secondary to lipoatrophy such as in individuals with generalized lipodystrophy. Therapies that restore leptin-LEPR signaling may resolve these metabolic sequelae. We developed a fully human monoclonal antibody (mAb), REGN4461 (mibavademab), that activates the human LEPR in the absence or presence of leptin. In obese leptin knockout mice, REGN4461 normalized body weight, food intake, blood glucose, and insulin sensitivity. In a mouse model of generalized lipodystrophy, REGN4461 alleviated hyperphagia, hyperglycemia, insulin resistance, dyslipidemia, and hepatic steatosis. In a phase 1, randomized, double-blind, placebo-controlled two-part study, REGN4461 was well tolerated with an acceptable safety profile. Treatment of individuals with overweight or obesity with REGN4461 decreased body weight over 12 weeks in those with low circulating leptin concentrations (<8 ng/ml) but had no effect on body weight in individuals with higher baseline leptin. Furthermore, compassionate-use treatment of a single patient with atypical partial lipodystrophy and a history of undetectable leptin concentrations associated with neutralizing antibodies to metreleptin was associated with noteable improvements in circulating triglycerides and hepatic steatosis. Collectively, these translational data unveil an agonist LEPR mAb that may provide clinical benefit in disorders associated with relatively low leptin concentrations.


Asunto(s)
Resistencia a la Insulina , Lipodistrofia Generalizada Congénita , Animales , Ratones , Humanos , Leptina/uso terapéutico , Ensayos de Uso Compasivo , Receptores de Leptina/metabolismo , Lipodistrofia Generalizada Congénita/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Anticuerpos/uso terapéutico , Peso Corporal
19.
Front Endocrinol (Lausanne) ; 14: 1257671, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37850099

RESUMEN

Introduction: Leptin inhibits insulin secretion from isolated islets from multiple species, but the cell type that mediates this process remains elusive. Several mouse models have been used to explore this question. Ablation of the leptin receptor (Lepr) throughout the pancreatic epithelium results in altered glucose homeostasis and ex vivo insulin secretion and Ca2+ dynamics. However, Lepr removal from neither alpha nor beta cells mimics this result. Moreover, scRNAseq data has revealed an enrichment of LEPR in human islet delta cells. Methods: We confirmed LEPR upregulation in human delta cells by performing RNAseq on fixed, sorted beta and delta cells. We then used a mouse model to test whether delta cells mediate the diminished glucose-stimulated insulin secretion in response to leptin. Results: Ablation of Lepr within mouse delta cells did not change glucose homeostasis or insulin secretion, whether mice were fed a chow or high-fat diet. We further show, using a publicly available scRNAseq dataset, that islet cells expressing Lepr lie within endothelial cell clusters. Conclusions: In mice, leptin does not influence beta-cell function through delta cells.


Asunto(s)
Insulina , Leptina , Animales , Humanos , Ratones , Glucosa/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Transducción de Señal
20.
J Endocrinol ; 259(3)2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37902096

RESUMEN

Absence of leptin confers metabolic dysfunction resulting in morbid obesity. Bone growth and maturation are also impaired. Partial leptin resistance is more common than leptin deficiency and, when induced by feeding mice a high fat diet, often has a negative effect on bone. Here, we used a genetic model to investigate the skeletal effects of partial and total leptin resistance in mice. This was accomplished by comparing the skeletal phenotypes of 17-week-old female C57Bl6/J wild-type (WT) mice, partial leptin receptor-deficient (db/+) mice and leptin receptor-deficient (db/db) mice (n = 7-8/group), all fed a standard diet. Compared to WT mice, db/db mice were dramatically heavier and hyperleptinemic. These mice were also hypogonadal, hyperglycemic, osteopenic and had lower serum levels of bone turnover markers, osteocalcin and C-terminal telopeptide of type I collagen (CTX). Compared to WT mice, db/+ mice were 14% heavier, had 149% more abdominal white adipose tissue, and were mildly hyperglycemic. db/+ mice did not differ from WT mice in uterine weight or serum levels of markers of bone turnover, although there was a trend for lower osteocalcin. At the bone microarchitectural level, db/+ mice differed from WT mice in having more massive femurs and a trend (P = 0.072) for larger vertebrae. These findings suggest that db/+ mice fed a normal mouse diet compensate for partial leptin resistance by increasing white adipose tissue mass which results in higher leptin levels. Our findings suggest that db/+ mice are a useful diet-independent model for studying the effects of partial leptin resistance on the skeleton.


Asunto(s)
Leptina , Receptores de Leptina , Femenino , Ratones , Animales , Leptina/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Osteocalcina/genética , Huesos/metabolismo , Dieta Alta en Grasa/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA