Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Diabetes Obes Metab ; 26(6): 2368-2378, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38560764

RESUMEN

AIM: To describe the biomarker strategy that was applied to select survodutide (BI 456906), BI 456908 and BI 456897 from 19 dual glucagon receptor (GCGR)/ glucagon-like peptide-1 receptor (GLP-1R) agonists for in-depth pharmacological profiling, which led to the qualification of survodutide as the clinical development candidate. MATERIALS AND METHODS: Potencies to increase cyclic adenosine monophosphate (cAMP) were determined in Chinese hamster ovary (CHO)-K1 cells stably expressing human GCGR and GLP-1R. Agonism for endogenously expressed receptors was investigated in insulinoma cells (MIN6) for mouse GLP-1R, and in rat primary hepatocytes for the GCGR. In vivo potencies to engage the GLP-1R or GCGR were determined, measuring improvement in oral glucose tolerance (30 nmol/kg) and increase in plasma fibroblast growth factor-21 (FGF21) and liver nicotinamide N-methyltransferase (NNMT) mRNA expression (100 nmol/kg), respectively. Body weight- and glucose-lowering efficacies were investigated in diet-induced obese (DIO) mice and diabetic db/db mice, respectively. RESULTS: Upon acute dosing in lean mice, target engagement biomarkers for the GCGR and GLP-1R demonstrated a significant correlation (Spearman correlation coefficient with p < 0.05) to the in vitro GCGR and GLP-1R potencies for the 19 dual agonists investigated. Survodutide, BI 456908 and BI 456897 were selected for in-depth pharmacological profiling based on the significant improvement in acute oral glucose tolerance achieved (area under the curve [AUC] of 54%, 57% and 60% vs. vehicle) that was comparable to semaglutide (AUC of 45% vs. vehicle), while showing different degrees of in vivo GCGR engagement, as determined by hepatic NNMT mRNA expression (increased by 15- to 17-fold vs. vehicle) and plasma FGF21 concentrations (increased by up to sevenfold vs. vehicle). In DIO mice, survodutide (30 nmol/kg/once daily), BI 456908 (30 nmol/kg/once daily) and BI 456897 (10 nmol/kg/once daily) achieved a body weight-lowering efficacy from baseline of 25%, 27% and 26%, respectively. In db/db mice, survodutide and BI 456908 (10 and 20 nmol/kg/once daily) significantly lowered glycated haemoglobin (0.4%-0.6%); no significant effect was observed for BI 456897 (3 and 7 nmol/kg/once daily). CONCLUSIONS: Survodutide was selected as the clinical candidate based on its balanced dual GCGR/GLP-1R pharmacology, engaging the GCGR for robust body weight-lowering efficacy exceeding that of selective GLP-1R agonists, while achieving antidiabetic efficacy that was comparable to selective GLP-1R agonism. Survodutide is currently being investigated in Phase 3 clinical trials in people living with obesity.


Asunto(s)
Cricetulus , Receptor del Péptido 1 Similar al Glucagón , Hipoglucemiantes , Receptores de Glucagón , Animales , Receptores de Glucagón/agonistas , Receptores de Glucagón/genética , Ratones , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/genética , Células CHO , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Humanos , Biomarcadores/sangre , Masculino , Ratas , Ratones Obesos , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico
2.
STAR Protoc ; 4(4): 102597, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37740914

RESUMEN

Immune dysregulation and inflammation by hepatic-resident leukocytes is considered a key step in disease progression of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis toward cirrhosis and hepatocellular carcinoma. Here, we provide a protocol for isolation and characterization of liver-resident immune cells from fine-needle biopsies obtained from a rodent model and humans. We describe steps for isolating leukocytes, cell sorting, and RNA extraction and sequencing. We then detail procedures for low-input mRNA sequencing analyses.


Asunto(s)
Neoplasias Hepáticas , Enfermedad del Hígado Graso no Alcohólico , Humanos , Ratones , Animales , Enfermedad del Hígado Graso no Alcohólico/patología , Cirrosis Hepática/patología , Biopsia
3.
Nat Commun ; 14(1): 4162, 2023 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-37443109

RESUMEN

The current obesity epidemic and high prevalence of metabolic diseases necessitate efficacious and safe treatments. Brown adipose tissue in this context is a promising target with the potential to increase energy expenditure, however no pharmacological treatments activating brown adipose tissue are currently available. Here, we identify AXL receptor tyrosine kinase as a regulator of adipose function. Pharmacological and genetic inhibition of AXL enhance thermogenic capacity of brown and white adipocytes, in vitro and in vivo. Mechanistically, these effects are mediated through inhibition of PI3K/AKT/PDE signaling pathway, resulting in induction of nuclear FOXO1 localization and increased intracellular cAMP levels via PDE3/4 inhibition and subsequent stimulation of the PKA-ATF2 pathway. In line with this, both constitutive Axl deletion as well as inducible adipocyte-specific Axl deletion protect animals from diet-induced obesity concomitant with increases in energy expenditure. Based on these data, we propose AXL receptor as a target for the treatment of obesity.


Asunto(s)
Tejido Adiposo Pardo , Tirosina Quinasa del Receptor Axl , Ratones , Animales , Tejido Adiposo Pardo/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Obesidad/metabolismo , Adipocitos Blancos/metabolismo , Metabolismo Energético , Tejido Adiposo Blanco/metabolismo , Termogénesis/genética , Adipocitos Marrones/metabolismo , Ratones Endogámicos C57BL , Tejido Adiposo/metabolismo
4.
Peptides ; 167: 171047, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37328068

RESUMEN

The number of diabetic patients is rising globally and concomitantly so do the diabetes associated complications. The gut secretes a variety of proteins to control blood glucose levels and/or food intake. As the drug class of GLP-1 agonists is based on a gut secreted peptide and the positive metabolic effects of bariatric surgery are at least partially mediated by gut peptides, we were interested in other gut secreted proteins which have yet to be explored. In this respect we identified the gut secreted protein FAM3D by analyzing sequencing data from L- and epithelial cells of VSG and sham operated as well as chow and HFD fed mice. FAM3D was overexpressed in diet induced obese mice via an adeno-associated virus (AAV), which resulted in a significant improvement of fasting blood glucose levels, glucose tolerance and insulin sensitivity. The liver lipid deposition was reduced, and the steatosis morphology was improved. Hyperinsulinemic clamps indicated that FAM3D is a global insulin sensitizer and increases glucose uptake into various tissues. In conclusion, the current study demonstrated that FAM3D controls blood glucose levels by acting as an insulin sensitizing protein and improves hepatic lipid deposition.


Asunto(s)
Hígado Graso , Resistencia a la Insulina , Ratones , Animales , Glucemia/metabolismo , Insulina/metabolismo , Hígado/metabolismo , Hígado Graso/metabolismo , Péptidos/farmacología , Lípidos , Ratones Endogámicos C57BL , Glucosa/metabolismo , Dieta Alta en Grasa , Citocinas/metabolismo
5.
Mol Metab ; 66: 101633, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36356832

RESUMEN

OBJECTIVE: Obesity and its associated comorbidities represent a global health challenge with a need for well-tolerated, effective, and mechanistically diverse pharmaceutical interventions. Oxyntomodulin is a gut peptide that activates the glucagon receptor (GCGR) and glucagon-like peptide-1 receptor (GLP-1R) and reduces bodyweight by increasing energy expenditure and reducing energy intake in humans. Here we describe the pharmacological profile of the novel glucagon receptor (GCGR)/GLP-1 receptor (GLP-1R) dual agonist BI 456906. METHODS: BI 456906 was characterized using cell-based in vitro assays to determine functional agonism. In vivo pharmacological studies were performed using acute and subchronic dosing regimens to demonstrate target engagement for the GCGR and GLP-1R, and weight lowering efficacy. RESULTS: BI 456906 is a potent, acylated peptide containing a C18 fatty acid as a half-life extending principle to support once-weekly dosing in humans. Pharmacological doses of BI 456906 provided greater bodyweight reductions in mice compared with maximally effective doses of the GLP-1R agonist semaglutide. BI 456906's superior efficacy is the consequence of increased energy expenditure and reduced food intake. Engagement of both receptors in vivo was demonstrated via glucose tolerance, food intake, and gastric emptying tests for the GLP-1R, and liver nicotinamide N-methyltransferase mRNA expression and circulating biomarkers (amino acids, fibroblast growth factor-21) for the GCGR. The dual activity of BI 456906 at the GLP-1R and GCGR was supported using GLP-1R knockout and transgenic reporter mice, and an ex vivo bioactivity assay. CONCLUSIONS: BI 456906 is a potent GCGR/GLP-1R dual agonist with robust anti-obesity efficacy achieved by increasing energy expenditure and decreasing food intake.


Asunto(s)
Péptido 1 Similar al Glucagón , Receptores de Glucagón , Animales , Humanos , Ratones , Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Oxintomodulina/farmacología , Péptidos/farmacología , Péptidos/metabolismo , Receptores de Glucagón/metabolismo
6.
Front Cell Dev Biol ; 10: 910040, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36092714

RESUMEN

The therapeutic potential of pluripotent stem cells is great as they promise to usher in a new era of medicine where cells or organs may be prescribed to replace dysfunctional tissue. At the forefront are efforts in the eye to develop this technology as it lends itself to in vivo monitoring and sophisticated non-invasive imaging modalities. In the retina, retinal pigment epithelium (RPE) is the most promising replacement cell as it has a single layer, is relatively simple to transplant, and is associated with several eye diseases. However, after transplantation, the cells may transform and cause complications. This transformation may be partially due to incomplete maturation. With the goal of learning how to mature RPE, we compared induced pluripotent stem cell-derived RPE (iPSC-RPE) cells with adult human primary RPE (ahRPE) cells and the immortalized human ARPE-19 line. We cultured ARPE-19, iPSC-RPE, and ahRPE cells for one month, and evaluated morphology, RPE marker staining, and transepithelial electrical resistance (TEER) as quality control indicators. We then isolated RNA for bulk RNA-sequencing and DNA for genotyping. We genotyped ahRPE lines for the top age-related macular degeneration (AMD) and proliferative vitreoretinopathy (PVR) risk allele polymorphisms. Transcriptome data verified that both adult and iPSC-RPE exhibit similar RPE gene expression signatures, significantly higher than ARPE-19. In addition, in iPSC-RPE, genes relating to stem cell maintenance, retina development, and muscle contraction were significantly upregulated compared to ahRPE. We compared ahRPE to iPSC-RPE in a model of epithelial-mesenchymal transition (EMT) and observed an increased sensitivity of iPSC-RPE to producing contractile aggregates in vitro which resembles incident reports upon transplantation. P38 inhibition was capable of inhibiting iPSC-RPE-derived aggregates. In summary, we find that the transcriptomic signature of iPSC-RPE conveys an immature RPE state which may be ameliorated by targeting "immature" gene regulatory networks.

7.
Nat Metab ; 4(1): 90-105, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35027768

RESUMEN

Healthy adipose tissue remodeling depends on the balance between de novo adipogenesis from adipogenic progenitor cells and the hypertrophy of adipocytes. De novo adipogenesis has been shown to promote healthy adipose tissue expansion, which confers protection from obesity-associated insulin resistance. Here, we define the role and trajectory of different adipogenic precursor subpopulations and further delineate the mechanism and cellular trajectory of adipogenesis, using single-cell RNA-sequencing datasets of murine adipogenic precursors. We identify Rspo2 as a functional regulator of adipogenesis, which is secreted by a subset of CD142+ cells to inhibit maturation of early progenitors through the receptor Lgr4. Increased circulating RSPO2 in mice leads to adipose tissue hypertrophy and insulin resistance and increased RSPO2 levels in male obese individuals correlate with impaired glucose homeostasis. Taken together, these findings identify a complex cellular crosstalk that inhibits adipogenesis and impairs adipose tissue homeostasis.


Asunto(s)
Adipogénesis , Tejido Adiposo/metabolismo , Redes y Vías Metabólicas , Trombospondinas/genética , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Tejido Adiposo/citología , Animales , Biología Computacional , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Heterogeneidad Genética , Humanos , Inmunofenotipificación , Resistencia a la Insulina , Ratones , Obesidad/etiología , Obesidad/metabolismo , RNA-Seq , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes , Células Madre/citología , Células Madre/metabolismo , Trombospondinas/metabolismo
8.
Nat Commun ; 12(1): 7144, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34880217

RESUMEN

Activation of thermogenic brown and beige adipocytes is considered as a strategy to improve metabolic control. Here, we identify GPR180 as a receptor regulating brown and beige adipocyte function and whole-body glucose homeostasis, whose expression in humans is associated with improved metabolic control. We demonstrate that GPR180 is not a GPCR but a component of the TGFß signalling pathway and regulates the activity of the TGFß receptor complex through SMAD3 phosphorylation. In addition, using genetic and pharmacological tools, we provide evidence that GPR180 is required to manifest Collagen triple helix repeat containing 1 (CTHRC1) action to regulate brown and beige adipocyte activity and glucose homeostasis. In this work, we show that CTHRC1/GPR180 signalling integrates into the TGFß signalling as an alternative axis to fine-tune and achieve low-grade activation of the pathway to prevent pathophysiological response while contributing to control of glucose and energy metabolism.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Termogénesis/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Adipocitos Beige/metabolismo , Adipocitos Marrones/metabolismo , Animales , Metabolismo Energético , Proteínas de la Matriz Extracelular/genética , Glucosa , Homeostasis , Humanos , Masculino , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Síndrome Metabólico/genética , Síndrome Metabólico/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/genética , Termogénesis/genética
9.
Nat Med ; 27(11): 1928-1940, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34663987

RESUMEN

Genes involved in distinct diabetes types suggest shared disease mechanisms. Here we show that One Cut Homeobox 1 (ONECUT1) mutations cause monogenic recessive syndromic diabetes in two unrelated patients, characterized by intrauterine growth retardation, pancreas hypoplasia and gallbladder agenesis/hypoplasia, and early-onset diabetes in heterozygous relatives. Heterozygous carriers of rare coding variants of ONECUT1 define a distinctive subgroup of diabetic patients with early-onset, nonautoimmune diabetes, who respond well to diabetes treatment. In addition, common regulatory ONECUT1 variants are associated with multifactorial type 2 diabetes. Directed differentiation of human pluripotent stem cells revealed that loss of ONECUT1 impairs pancreatic progenitor formation and a subsequent endocrine program. Loss of ONECUT1 altered transcription factor binding and enhancer activity and NKX2.2/NKX6.1 expression in pancreatic progenitor cells. Collectively, we demonstrate that ONECUT1 controls a transcriptional and epigenetic machinery regulating endocrine development, involved in a spectrum of diabetes, encompassing monogenic (recessive and dominant) as well as multifactorial inheritance. Our findings highlight the broad contribution of ONECUT1 in diabetes pathogenesis, marking an important step toward precision diabetes medicine.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Factor Nuclear 6 del Hepatocito/genética , Páncreas/embriología , Diferenciación Celular/genética , Anomalías Congénitas/genética , Retardo del Crecimiento Fetal/genética , Vesícula Biliar/anomalías , Proteína Homeobox Nkx-2.2/biosíntesis , Proteínas de Homeodominio/biosíntesis , Humanos , Lactante , Recién Nacido , Masculino , Herencia Multifactorial/genética , Organogénesis/genética , Páncreas/anomalías , Enfermedades Pancreáticas/congénito , Enfermedades Pancreáticas/genética , Células Madre Pluripotentes/citología , Transcripción Genética/genética
10.
Pharmaceutics ; 13(8)2021 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-34452066

RESUMEN

Rho-associated kinase (ROCK) activation was shown to contribute to microvascular closure, retinal hypoxia, and to retinal pigment epithelium (RPE) barrier disruption in a rat model of diabetic retinopathy. Fasudil, a clinically approved ROCK inhibitor, improved retinal perfusion and reduced edema in this model, indicating that ROCK inhibition could be a promising new therapeutic approach for the treatment of diabetic retinopathy. However, due to its short intravitreal half-life, fasudil is not suitable for long-term treatment. In this study, we evaluated a very potent ROCK1/2 inhibitor (BIRKI) in a depot formulation administered as a single intravitreal injection providing a slow release for at least four weeks. Following BIRKI intravitreal injection in old Goto-Kakizaki (GK) type 2 diabetic rats, we observed a significant reduction in ROCK1 activity in the retinal pigment epithelium/choroid complex after 8 days and relocation of ROCK1 to the cytoplasm and nucleus in retinal pigment epithelium cells after 28 days. The chronic ROCK inhibition by the BIRKI depot formulation restored retinal pigment epithelial cell morphology and distribution, favored retinal capillaries dilation, and reduced hypoxia and inner blood barrier leakage observed in the diabetic retina. No functional or morphological negative effects were observed, indicating suitable tolerability of BIRKI after intravitreous injection. In conclusion, our data suggest that sustained ROCK inhibition, provided by BIRKI slow-release formulation, could be a valuable treatment option for diabetic retinopathy, especially with regard to the improvement of retinal vascular infusion and protection of the outer retinal barrier.

11.
Mol Metab ; 51: 101237, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33878401

RESUMEN

BACKGROUND: A chronic imbalance of energy intake and energy expenditure results in excess fat storage. The obesity often caused by this overweight is detrimental to the health of millions of people. Understanding both sides of the energy balance equation and their counter-regulatory mechanisms is critical to the development of effective therapies to treat this epidemic. SCOPE OF REVIEW: Behaviors surrounding ingestion have been reviewed extensively. This review focuses more specifically on energy expenditure regarding bodyweight control, with a particular emphasis on the organs and attractive metabolic processes known to reduce bodyweight. Moreover, previous and current attempts at anti-obesity strategies focusing on energy expenditure are highlighted. Precise measurements of energy expenditure, which consist of cellular, animal, and human models, as well as measurements of their translatability, are required to provide the most effective therapies. MAJOR CONCLUSIONS: A precise understanding of the components surrounding energy expenditure, including tailored approaches based on genetic, biomarker, or physical characteristics, must be integrated into future anti-obesity treatments. Further comprehensive investigations are required to define suitable treatments, especially because the complex nature of the human perspective remains poorly understood.


Asunto(s)
Ingestión de Energía , Metabolismo Energético/fisiología , Obesidad/terapia , Animales , Modelos Animales de Enfermedad , Humanos , Obesidad/metabolismo , Obesidad/fisiopatología
12.
Commun Chem ; 3(1): 75, 2020 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-36703453

RESUMEN

The C-type lectin family member lectin-like oxidized LDL receptor-1 (LOX-1) has been object of intensive research. Its modulation may offer a broad spectrum of therapeutic interventions ranging from cardiovascular diseases to cancer. LOX-1 mediates uptake of oxLDL by vascular cells and plays an important role in the initiation of endothelial dysfunction and its progression to atherosclerosis. So far only a few compounds targeting oxLDL-LOX-1 interaction are reported with a limited level of characterization. Here we describe the identification and characterization of BI-0115, a selective small molecule inhibitor of LOX-1 that blocks cellular uptake of oxLDL. Identified by a high throughput screening campaign, biophysical analysis shows that BI-0115 binding triggers receptor inhibition by formation of dimers of the homodimeric ligand binding domain. The structure of LOX-1 bound to BI-0115 shows that inter-ligand interactions at the receptor interfaces are key to the formation of the receptor tetramer thereby blocking oxLDL binding.

13.
J Endocrinol ; 244(2): 323-337, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31682591

RESUMEN

Maturity-onset diabetes of the young (MODY) is a group of monogenetic forms of diabetes mellitus caused by mutations in genes regulating ß-cell development and function. MODY represents a heterogeneous group of non-insulin-dependent diabetes arising in childhood or adult life. Interestingly, clinical heterogeneity in MODY patients like variable disease onset and severity is observed even among individual family members sharing the same mutation, an issue that is not well understood. As high blood glucose levels are a well-known factor promoting ß-cell stress and ultimately leading to cell death, we asked whether additional ß-cell stress might account for the occurrence of disease heterogeneity in mice carrying a MODY4 mutation. In order to challenge ß-cells, we established a MODY4 animal model based on Pdx1 (pancreatic and duodenal homeobox 1) haploinsufficiency, which allows conditional modulation of cell stress by genetic inhibition of the stress-responsive IKK/NF-κB signalling pathway. While Pdx1+/- mice were found glucose intolerant without progressing to diabetes, additional challenge of ß-cell function by IKK/NF-κB inhibition promoted rapid diabetes development showing hyperglycaemia, hypoinsulinemia and loss of ß-cell mass. Disease pathogenesis was characterized by deregulation of genes controlling ß-cell homeostasis and function. Importantly, restoration of normal IKK/NF-κB signalling reverted the diabetic phenotype including normalization of glycaemia and ß-cell mass. Our findings implicate that the avoidance of additional ß-cell stress can delay a detrimental disease progression in MODY4 diabetes. Remarkably, an already present diabetic phenotype can be reversed when ß-cell stress is normalized.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Glucemia/metabolismo , Muerte Celular , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/metabolismo , Estrés Fisiológico , Transactivadores/genética , Transactivadores/metabolismo
14.
Mol Metab ; 23: 51-59, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30905616

RESUMEN

OBJECTIVE: Roux-en-Y gastric bypass (RYGB) surgery produces rapid and persistent reductions in plasma triglyceride (TG) levels associated with fewer cardiovascular events. The mechanisms of the reduction in systemic TG levels remain unclear. We hypothesized that RYGB reduces intestinal TG secretion via altered enterocyte lipid handling. METHODS: RYGB or Sham surgery was performed in diet-induced obese, insulin-resistant male Sprague-Dawley rats. First, we tested whether RYGB reduced test meal-induced TG levels in the intestinal lymph, a direct readout of enterocyte lipid secretion. Second, we examined whether RYGB modified TG enterocyte secretion at the single lipid level and in comparison to other lipid subclasses, applying mass spectrometry lipidomics to the intestinal lymph of RYGB and Sham rats (0-21 days after surgery). Third, we explored whether RYGB modulated the metabolic characteristics of primary enterocytes using transcriptional and functional assays relevant to TG absorption, reesterification, storage in lipid droplets, and oxidation. RESULTS: RYGB reduced overall postprandial TG concentrations compared to Sham surgery in plasma and intestinal lymph similarly. RYGB reduced lymphatic TG concentrations more than other lipid subclasses, and shifted the remaining TG pool towards long-chain, unsaturated species. In enterocytes of fasted RYGB rats, lipid uptake was transcriptionally (Fatp4, Fabp2, Cd36) and functionally reduced compared to Sham, whereas TG reesterification genes were upregulated. CONCLUSION: Our results show that RYGB substantially reduces intestinal TG secretion and modifies enterocyte lipid absorption and handling in rats. These changes likely contribute to the improvements in the plasma TG profile observed after RYGB in humans.


Asunto(s)
Enterocitos/metabolismo , Derivación Gástrica/métodos , Periodo Posprandial/fisiología , Triglicéridos/sangre , Animales , Glucemia , Diglicéridos/metabolismo , Modelos Animales de Enfermedad , Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina , Mucosa Intestinal/metabolismo , Linfa/metabolismo , Masculino , Obesidad/cirugía , Ratas , Ratas Sprague-Dawley
15.
Peptides ; 113: 22-34, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30660763

RESUMEN

Altered GLP-1 secretion from L cells has been implicated in the development of type 2 diabetes mellitus and its resolution following bariatric surgery. However, changes in L cell gene expression, which may form the basis for altered functionality after high fat diet (HFD) or bariatric surgery, have either not been investigated or have given conflicting results. We developed a gcg-DTR-eGFP reporter mouse to isolate ileal and colonic L cells from HFD fed insulin resistant mice and mice showing improved glucose tolerance following vertical sleeve gastrectomy (VSG). Transcriptomic sequencing and identification of genes differentially expressed in response to HFD or VSG revealed small changes with HFD, primarily in immune related genes, but no regulation following VSG. In contrast, large differences were observed between ileal and colonic L cells due to the differential expression of genes involved in nutrient transport and metabolism, reflecting to some extent the differences in the surrounding epithelium. We showed that, in line with the gene expression data, colonic and ileal L cells exhibit differing GLP-1 responses to nutrients (glucose and the gly-sar dipeptide) and hormones (vasopressin). Thus, we hypothesise that colonic and ileal L cells have different physiological roles, with ileal L cells contributing more to postprandial glucose homeostasis by responding to dietary nutrients and colonic cells responding more to non-dietary stimulants.


Asunto(s)
Glucemia/metabolismo , Colon/metabolismo , Gastrectomía/efectos adversos , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/metabolismo , Íleon/metabolismo , Animales , Cirugía Bariátrica , Dieta Alta en Grasa , Homeostasis , Masculino , Ratones , Periodo Posprandial , Análisis de Secuencia de ARN , Transcriptoma
16.
Cell Rep ; 25(3): 784-797.e4, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30332656

RESUMEN

Recruitment and activation of thermogenic adipocytes have received increasing attention as a strategy to improve systemic metabolic control. The analysis of brown and brite adipocytes is complicated by the complexity of adipose tissue biopsies. Here, we provide an in-depth analysis of pure brown, brite, and white adipocyte transcriptomes. By combining mouse and human transcriptome data, we identify a gene signature that can classify brown and white adipocytes in mice and men. Using a machine-learning-based cell deconvolution approach, we develop an algorithm proficient in calculating the brown adipocyte content in complex human and mouse biopsies. Applying this algorithm, we can show in a human weight loss study that brown adipose tissue (BAT) content is associated with energy expenditure and the propensity to lose weight. This online available tool can be used for in-depth characterization of complex adipose tissue samples and may support the development of therapeutic strategies to increase energy expenditure in humans.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Biomarcadores/análisis , Biología Computacional/métodos , Obesidad/fisiopatología , Programas Informáticos , Adipogénesis , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Adulto , Anciano , Animales , Estudios de Cohortes , Metabolismo Energético , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Termogénesis , Adulto Joven
17.
Sci Signal ; 10(466)2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28196906

RESUMEN

Brown adipose tissue (BAT) is responsible for thermogenesis that is not associated with shivering through the process of converting chemical energy into heat through uncoupling protein 1 (UCP1) in the mitochondria. Thus, expanding or activating BAT could be a potential tool against obesity. To analyze the effect of kinase signaling on brown adipocyte formation, a process that describes the acquisition of the ability to dissipate energy as heat, we performed lentiviral-mediated short hairpin knockdown or used pharmacological inhibitors in a high-content and high-throughput in vitro image-based screen. We identified 190 kinases that either stimulated or inhibited brown adipocyte proliferation, differentiation, or formation. Among these kinases, we found that 5' AMP-activated protein kinase (AMPK) promoted the formation of brown adipocytes abundant inUCP1. Together, our results provide insight into the kinases, particularly AMPK, that regulate brown adipocyte formation.


Asunto(s)
Adipocitos Marrones/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Fosfotransferasas/genética , Interferencia de ARN , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos Marrones/citología , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Animales , Western Blotting , Células Cultivadas , Ratones , Fosfotransferasas/clasificación , Fosfotransferasas/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteómica/métodos
18.
Cell Rep ; 16(8): 2243-2258, 2016 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-27524617

RESUMEN

While Bmp4 has a well-established role in the commitment of mesenchymal stem cells into the adipogenic lineage, its role in brown adipocyte formation and activity is not well defined. Here, we show that Bmp4 has a dual function in adipogenesis by inducing adipocyte commitment while inhibiting the acquisition of a brown phenotype during terminal differentiation. Selective brown adipose tissue overexpression of Bmp4 in mice induces a shift from a brown to a white-like adipocyte phenotype. This effect is mediated by Smad signaling and might be in part due to suppression of lipolysis, via regulation of hormone sensitive lipase expression linked to reduced Ppar activity. Given that we observed a strong correlation between BMP4 levels and adipocyte size, as well as insulin sensitivity in humans, we propose that Bmp4 is an important factor in the context of obesity and type 2 diabetes.


Asunto(s)
Adipocitos Marrones/efectos de los fármacos , Adipocitos Blancos/efectos de los fármacos , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Proteína Morfogenética Ósea 4/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Adipocitos Marrones/citología , Adipocitos Marrones/metabolismo , Adipocitos Blancos/citología , Adipocitos Blancos/metabolismo , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/metabolismo , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular , Línea Celular Transformada , AMP Cíclico/farmacología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Regulación de la Expresión Génica , Humanos , Resistencia a la Insulina , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores Activados del Proliferador del Peroxisoma/genética , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Rosiglitazona , Transducción de Señal , Proteínas Smad/genética , Proteínas Smad/metabolismo , Esterol Esterasa/genética , Esterol Esterasa/metabolismo , Tiazolidinedionas/farmacología
19.
Mol Metab ; 4(11): 795-810, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26629404

RESUMEN

OBJECTIVE: Failure to properly dispose of glucose in response to insulin is a serious health problem, occurring during obesity and is associated with type 2 diabetes development. Insulin-stimulated glucose uptake is facilitated by the translocation and plasma membrane fusion of vesicles containing glucose transporter 4 (GLUT4), the rate-limiting step of post-prandial glucose disposal. METHODS: We analyzed the role of Tusc5 in the regulation of insulin-stimulated Glut4-mediated glucose uptake in vitro and in vivo. Furthermore, we measured Tusc5 expression in two patient cohorts. RESULTS: Herein, we report that TUSC5 controls insulin-stimulated glucose uptake in adipocytes, in vitro and in vivo. TUSC5 facilitates the proper recycling of GLUT4 and other key trafficking proteins during prolonged insulin stimulation, thereby enabling proper protein localization and complete vesicle formation, processes that ultimately enable insulin-stimulated glucose uptake. Tusc5 knockout mice exhibit impaired glucose disposal and TUSC5 expression is predictive of glucose tolerance in obese individuals, independent of body weight. Furthermore, we show that TUSC5 is a PPARγ target and in its absence the anti-diabetic effects of TZDs are significantly blunted. CONCLUSIONS: Collectively, these findings establish TUSC5 as an adipose tissue-specific protein that enables proper protein recycling, linking the ubiquitous vesicle traffic machinery with tissue-specific insulin-mediated glucose uptake into adipose tissue and the maintenance of a healthy metabolic phenotype in mice and humans.

20.
PLoS One ; 9(12): e116288, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25551639

RESUMEN

In recent years, the increasing prevalence of obesity and obesity-related co-morbidities fostered intensive research in the field of adipose tissue biology. To further unravel molecular mechanisms of adipose tissue function, genetic tools enabling functional studies in vitro and in vivo are essential. While the use of transgenic animals is well established, attempts using viral and non-viral vectors to genetically modify adipocytes in vivo are rare. Therefore, we here characterized recombinant Adeno-associated virus (rAAV) vectors regarding their potency as gene transfer vehicles for adipose tissue. Our results demonstrate that a single dose of systemically applied rAAV8-CMV-eGFP can give rise to remarkable transgene expression in murine adipose tissues. Upon transcriptional targeting of the rAAV8 vector to adipocytes using a 2.2 kb fragment of the murine adiponectin (mAP2.2) promoter, eGFP expression was significantly decreased in off-target tissues while efficient transduction was maintained in subcutaneous and visceral fat depots. Moreover, rAAV8-mAP2.2-mediated expression of perilipin A - a lipid-droplet-associated protein - resulted in significant changes in metabolic parameters only three weeks post vector administration. Taken together, our findings indicate that rAAV vector technology is applicable as a flexible tool to genetically modify adipocytes for functional proof-of-concept studies and the assessment of putative therapeutic targets in vivo.


Asunto(s)
Tejido Adiposo/virología , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Adiponectina/genética , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Dependovirus/genética , Vectores Genéticos , Proteínas Fluorescentes Verdes/análisis , Ratones , Perilipina-1 , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...