375 results on '"insulin signaling"'
Search Results
2. Dietary soy prevents fetal demise, intrauterine growth restriction, craniofacial dysmorphic features, and impairments in placentation linked to gestational alcohol exposure: Pivotal role of insulin and insulin-like growth factor signaling networks.
- Author
-
Qi, Wei, Gundogan, Fusun, Gilligan, Jeffrey, and Monte, Suzanne de la
- Subjects
- *
SOMATOMEDIN , *FETAL growth retardation , *FETAL alcohol syndrome , *PRENATAL alcohol exposure , *PLACENTAL growth factor , *EMBRYO implantation , *PHYTOESTROGENS - Abstract
Prenatal alcohol exposure can impair placentation and cause intrauterine growth restriction (IUGR), fetal demise, and fetal alcohol spectrum disorder (FASD). Previous studies showed that ethanol's inhibition of placental insulin and insulin-like growth factor, type 1 (IGF-1) signaling compromises trophoblastic cell motility and maternal vascular transformation at the implantation site. Since soy isolate supports insulin responsiveness, we hypothesized that dietary soy could be used to normalize placentation and fetal growth in an experimental model of FASD. Pregnant Long-Evans rat dams were fed with isocaloric liquid diets containing 0% or 8.2% ethanol (v/v) from gestation day (GD) 6. Dietary protein sources were either 100% soy isolate or 100% casein (standard). Gestational sacs were harvested on GD19 to evaluate fetal resorption, fetal growth parameters, and placental morphology. Placental insulin/IGF-1 signaling through Akt pathways was assessed using commercial bead-based multiplex enzyme-linked immunosorbent assays. Dietary soy markedly reduced or prevented the ethanol-associated fetal loss, IUGR, FASD dysmorphic features, and impairments in placentation/maturation. Furthermore, ethanol's inhibitory effects on the placental glycogen cell population at the junctional zone, invasive trophoblast populations at the implantation site, maternal vascular transformation, and signaling through the insulin and IGF1 receptors, Akt and PRAS40 were largely abrogated by co-administration of soy. Dietary soy may provide an economically feasible and accessible means of reducing adverse pregnancy outcomes linked to gestational ethanol exposure. • Gestational dietary soy consumption prevents fetal loss, intrauterine growth restriction, and critical dysmorphic craniofacial features of fetal alcohol spectrum disorder. • Dietary soy isolate preserves the integrity of placentation despite continuous high-level gestational alcohol exposure. • Soy isolate preserves the integrity of placental insulin signal transduction through Akt cell survival and metabolic pathways in an experimental fetal alcohol spectrum disorder model. • Soy isolate enhances development in control animals, suggesting it may provide an accessible means to globally reduce rates of low birth weight. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
3. Lespedeza bicolor extract supplementation reduced hyperglycemia-induced skeletal muscle damage by regulation of AMPK/SIRT/PGC1α–related energy metabolism in type 2 diabetic mice.
- Author
-
Lee, Heaji, Kim, Sun Yeou, and Lim, Yunsook
- Subjects
- *
INFLAMMATION prevention , *MUSCULAR atrophy , *ENERGY metabolism , *PROTEIN kinases , *GLYCOSYLATED hemoglobin , *PROTEINS , *HYPERGLYCEMIA , *SKELETAL muscle , *ANIMAL experimentation , *HYPOGLYCEMIC agents , *CELL receptors , *TYPE 2 diabetes , *CELLULAR signal transduction , *MITOCHONDRIA , *OXIDATIVE stress , *CELL proliferation , *PLANT extracts , *MICE , *PHARMACODYNAMICS , *DISEASE complications - Abstract
Lespedeza bicolor (LB) is known to have antidiabetic activities; however, the underlying molecular mechanisms of LB in hyperglycemia-induced skeletal muscle damage is unclear. Inflammation and oxidative stress caused by type 2 diabetes mellitus (T2DM) not only contributes to insulin resistance, but also promotes muscle atrophy via decreased muscle protein synthesis and increased protein degradation, leading to frailty and sarcopenia. In this study, we hypothesized that LB extract (LBE) supplementatin has an ameliorative effect on hyperglycemia-induced skeletal muscle damage by activation of 5′ adenosine monophosphate-activated protein kinase (AMPK)/sirtuin (SIRT)/proliferator-activated receptor γ coactivator 1α (PGC1α)–associated energy metabolism in mice with T2DM. Diabetes was induced by a high-fat diet with a 2-time streptozotoxin injection (30 mg/kg body weight) in male C57BL/6J mice. After diabetes was induced (fasting blood glucose level ≥140 mg/dL), the mice were administered with LBE at a low dose (100 mg/kg/d) or high dose (250 mg/kg/d) by gavage for 12 weeks. LBE supplementation ameliorated glucose tolerance and hemoglobin A1c (%) in mice with T2DM. Moreover, LBE supplementation upregulated protein levels of insulin receptor subunit-1 and Akt accompanied by increased translocation of glucose transporter 4 in mice with T2DM. Furthermore, LBE increased mitochondrial biogenesis by activating SIRT1, SIRT3, SIRT4, and peroxisome PGC1α in diabetic skeletal muscle. Meanwhile, LBE supplementation reduced oxidative stress and inflammation in mice with T2DM. Taken together, the current study suggested that LBE could be a potential therapeutic to prevent skeletal muscle damage by regulation AMPK/SIRT/PGC1α–related energy metabolism in T2DM. Lespedeza bicolor (LB) contains many antioxidant compounds, including genistein, quercetin, daidzein, catechin, luteolin, and naringin. The natural compounds rich in LB extract (LBE) improved skeletal muscle energy metabolism with insulin signaling by regulation of 5′ adenosine monophosphate-activated protein kinase (AMPK)/sirtuin (SIRT)1/proliferator-activated receptor gamma coactivator-1 α (PGC1α) pathway in type 2 diabetes mellitus (T2DM) mice. Furthermore, LBE reduced oxidative stress and inflammation causing muscle protein degradation in T2DM mice. Consequently, LBE is expected to be a potential nutraceutical for alleviating hyperglycemia-induced skeletal muscle damage in T2DM. ROS, reactive oxygen species. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
4. MicroRNA-mediated mechanisms in whole grain consumption for preventing type 2 diabetes mellitus.
- Author
-
Qin, Mengyuan, Chen, Zenglong, Liu, Xuan, and Ren, Xin
- Abstract
Whole grains emerge preventive or ameliorative effects on type 2 diabetes mellitus as one of the staple foods with abundant polyphenols and dietary fiber. However, there is currently no systematic review elucidating the metabolic mechanisms associated with whole-grain regulation of blood glucose metabolism based on novel disease marker microRNAs. The present paper systematically reviewed the metabolic mechanisms of whole grain for blood glucose regulation based on microRNA. In addition to the conventional mechanisms, including insulin resistance, inflammation, oxidative stress, and insulin signaling, we focused on the potential mechanisms of whole grains and their phytochemicals in modulating type 2 diabetes mellitus through microRNA. Whole grains and their phytochemicals could mediate different miRNAs to improve diabetes by intervening with different targets, such as glycolipid metabolism, inflammation, oxidative stress and gut microbial. This work would enrich our understanding of whole grains improving type 2 diabetes mellitus and promote the consumption of whole grains. • Whole grains rich in phytochemicals are helpful in improving diabetes. • MiRNAs are considered as novel T2DM biomarkers to regulate T2DM. • Whole grains and their phytochemicals mediate miRNA to regulate T2DM. • Quality and quantity of evidence for microRNA in whole grain glucose regulation were assessed. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Monocarbonyl analogs of curcumin C66 and B2BrBC modulate oxidative stress, JNK activity, and pancreatic gene expression in rats with streptozotocin-induced diabetes.
- Author
-
Stojchevski, Radoslav, Velichkovikj, Sara, Bogdanov, Jane, Hadzi-Petrushev, Nikola, Mladenov, Mitko, Poretsky, Leonid, and Avtanski, Dimiter
- Subjects
- *
TYPE 1 diabetes , *CHEMICAL stability , *WESTERN immunoblotting , *STREPTOZOTOCIN , *LABORATORY rats - Abstract
[Display omitted] The pathogenesis of type 1 diabetes mellitus (T1DM) involves oxidative stress and inflammation. Curcumin, a natural polyphenolic compound found in turmeric, known to exhibit antioxidative and anti-inflammatory properties, is characterized by poor chemical stability, low bioavailability, and rapid metabolism. Monocarbonyl analogs of curcumin (MACs) with a structural absence of β-diketone and enhanced stability and bioavailability present a potential solution to the challenges associated with the use of curcumin. This study aimed to evaluate the effect of two MACs, C66 and B2BrBC, on oxidative stress markers, antioxidant enzyme activity, expression of diabetes-associated genes, and signaling pathway proteins in the context of T1DM. Streptozotocin (STZ)-induced male Wistar rats or rat pancreatic RIN-m cells were used for in vivo and in vitro experiments, respectively. C66 or B2BrBC were given either before or after STZ treatment. Oxidative stress markers and antioxidant enzyme activities were determined in various tissues. Expression of diabetes-associated genes was assessed using RT-qPCR, and the activity of signaling pathway proteins in the pancreas was determined through Western blot analysis. Treatment with C66 and B2BrBC significantly reduced oxidative stress markers and positively influenced antioxidant enzyme activities. Moreover, both compounds inhibited JNK activity in the pancreas while enhancing the expression of genes crucial for β-cell survival and glucose and redox homeostasis. The findings highlight the multifaceted potential of C66 and B2BrBC in ameliorating oxidative stress, influencing gene expression patterns linked to diabetes, and modulating key signaling pathways in the pancreas. The findings suggest that these compounds can potentially address diabetes-related pathological processes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Evaluating combined effects of chronic, low-dose exposures of cadmium (CLEC) and hyperglycemia on insulin signaling dysfunction in a hepatocellular model.
- Author
-
Kumar, Rahul and Gullapalli, Rama R.
- Subjects
- *
TYPE 2 diabetes , *HEAVY metals , *REACTIVE oxygen species , *METABOLIC disorders , *GENE expression - Abstract
The pathophysiological effects of chronic heavy metal exposures on human health remains uncertain. In this study, we developed a novel chronic, low-dose exposure of Cadmium (CLEC) model using the hepatocellular cell lines, HepG2 and HUH7. We modulated cell culture conditions to mimic human normoglycemic (5.6 mM) and hyperglycemic (15 mM) states with concomitant cadmium (Cd) exposures for 24 weeks. CLEC cells undergo non-trivial alterations in glucose signaling and metabolic characteristics within our model. We observe elevated baseline reactive oxygen species (ROS) production and decreased 2-NBDG uptake indicative of glucose metabolic dysfunction. Additionally, induction of metallothionein (MT) expression, increased activation of Akt signaling (via phosphorylation) and reduced IRS-2 protein expression are observed in CLEC cells. Cell line specific changes are observed with HepG2 showing a much higher MT gene induction compared to HUH7 cell line which impacts glucose metabolic dysfunction. Hyperglycemic culture conditions (representing type II diabetes) significantly modulate CLEC effects on cells. In conclusion, pathophysiologically relevant models of chronic heavy metal exposures are urgently needed to gain an in-depth, mechanistic understanding of the long-term impacts of toxic metals (e.g., Cd) on human metabolic health. [Display omitted] • We developed an invitro model to study chronic low-dose cadmium (CLEC) effects in variable glycemic conditions. • CLEC enhances reactive oxygen species (ROS) production synergistically with hyperglycemic culture conditions (15 mM). • CLEC paradigm reduces cellular glucose uptake, which is exacerbated by hyperglycemic conditions. • CLEC upregulates metallothionein (MT-1 and MT2) gene expression which is attenuated by simultaneous hyperglycemia. • CLEC paradigm dysregulates IRS2 and AKT signaling activity, leading to impaired insulin signaling. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Tissue-specific activation of insulin signaling as a potential target for obesity-related metabolic disorders.
- Author
-
Okuma, Hideyuki and Tsuchiya, Kyoichiro
- Subjects
- *
VASCULAR endothelial cells , *VASCULAR smooth muscle , *TYPE 2 diabetes , *INSULIN receptors , *INSULIN sensitivity , *INSULIN - Abstract
The incidence of obesity is rapidly increasing worldwide. Obesity-associated insulin resistance has long been established as a significant risk factor for obesity-related disorders such as type 2 diabetes and atherosclerosis. Insulin plays a key role in systemic glucose metabolism, with the liver, skeletal muscle, and adipose tissue as the major acting tissues. Insulin receptors and the downstream insulin signaling-related molecules are expressed in various tissues, including vascular endothelial cells, vascular smooth muscle cells, and monocytes/macrophages. In obesity, decreased insulin action is considered a driver for associated disorders. However, whether insulin action has a positive or negative effect on obesity-related disorders depends on the tissue in which it acts. While an enhancement of insulin signaling in the liver increases hepatic fat accumulation and exacerbates dyslipidemia, enhancement of insulin signaling in adipose tissue protects against obesity-related dysfunction of various organs by increasing the capacity for fat accumulation in the adipose tissue and inhibiting ectopic fat accumulation. Thus, this "healthy adipose tissue expansion" by enhancing insulin sensitivity in adipose tissue, but not in the liver, may be an effective therapeutic strategy for obesity-related disorders. To effectively address obesity-related metabolic disorders, the mechanisms of insulin resistance in various tissues of obese patients must be understood and drugs that enhance insulin action must be developed. In this article, we review the potential of interventions that enhance insulin signaling as a therapeutic strategy for obesity-related disorders, focusing on the molecular mechanisms of insulin action in each tissue. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Picalm, a novel regulator of GLUT4-trafficking in adipose tissue.
- Author
-
Gaugel, Jasmin, Haacke, Neele, Sehgal, Ratika, Jähnert, Markus, Jonas, Wenke, Hoffmann, Anne, Blüher, Matthias, Ghosh, Adhideb, Noé, Falko, Wolfrum, Christian, Tan, Joycelyn, Schürmann, Annette, Fazakerley, Daniel J., and Vogel, Heike
- Abstract
Picalm (phosphatidylinositol-binding clathrin assembly protein), a ubiquitously expressed clathrin-adapter protein, is a well-known susceptibility gene for Alzheimer's disease, but its role in white adipose tissue (WAT) function has not yet been studied. Transcriptome analysis revealed differential expression of Picalm in WAT of diabetes-prone and diabetes-resistant mice, hence we aimed to investigate the potential link between Picalm expression and glucose homeostasis, obesity-related metabolic phenotypes, and its specific role in insulin-regulated GLUT4 trafficking in adipocytes. Picalm expression and epigenetic regulation by microRNAs (miRNAs) and DNA methylation were analyzed in WAT of diabetes-resistant (DR) and diabetes-prone (DP) female New Zealand Obese (NZO) mice and in male NZO after time-restricted feeding (TRF) and alternate-day fasting (ADF). PICALM expression in human WAT was evaluated in a cross-sectional cohort and assessed before and after weight loss induced by bariatric surgery. siRNA-mediated knockdown of Picalm in 3T3-L1-cells was performed to elucidate functional outcomes on GLUT4-translocation as well as insulin signaling and adipogenesis. Picalm expression in WAT was significantly lower in DR compared to DP female mice, as well as in insulin-sensitive vs. resistant NZO males, and was also reduced in NZO males following TRF and ADF. Four miRNAs (let-7c, miR-30c, miR-335, miR-344) were identified as potential mediators of diabetes susceptibility-related differences in Picalm expression, while 11 miRNAs (including miR-23a, miR-29b, and miR-101a) were implicated in TRF and ADF effects. Human PICALM expression in adipose tissue was lower in individuals without obesity vs. with obesity and associated with weight-loss outcomes post-bariatric surgery. siRNA-mediated knockdown of Picalm in mature 3T3-L1-adipocytes resulted in amplified insulin-stimulated translocation of the endogenous glucose transporter GLUT4 to the plasma membrane and increased phosphorylation of Akt and Tbc1d4. Moreover, depleting Picalm before and during 3T3-L1 differentiation significantly suppressed adipogenesis, suggesting that Picalm may have distinct roles in the biology of pre- and mature adipocytes. Picalm is a novel regulator of GLUT4-translocation in WAT, with its expression modulated by both genetic predisposition to diabetes and dietary interventions. These findings suggest a potential role for Picalm in improving glucose homeostasis and highlight its relevance as a therapeutic target for metabolic disorders. • Diabetes-resistant and lean mice exhibit lower Picalm expression in adipose tissue. • In humans, PICALM is higher expressed in adipose tissue of obese individuals. • Intermittent fasting is sufficient to reduce Picalm expression via epigenetic mechanisms. • Picalm knockdown increases insulin-stimulated GLUT4-translocation in adipocytes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. PPARG in osteocytes controls cell bioenergetics and systemic energy metabolism independently of sclerostin levels in circulation.
- Author
-
Baroi, Sudipta, Czernik, Piotr J., Khan, Mohd Parvez, Letson, Joshua, Crowe, Emily, Chougule, Amit, Griffin, Patrick R., Rosen, Clifford J., and Lecka-Czernik, Beata
- Abstract
The skeleton is one of the largest organs in the body, wherein metabolism is integrated with systemic energy metabolism. However, the bioenergetic programming of osteocytes, the most abundant bone cells coordinating bone metabolism, is not well defined. Here, using a mouse model with partial penetration of an osteocyte-specific PPARG deletion, we demonstrate that PPARG controls osteocyte bioenergetics and their contribution to systemic energy metabolism independently of circulating sclerostin levels, which were previously correlated with metabolic status of extramedullary fat depots. In vivo and in vitro models of osteocyte-specific PPARG deletion, i.e. Dmp 1
Cre Pparγflfl male and female mice (γOTKO ) and MLO-Y4 osteocyte-like cells with either siRNA-silenced or CRISPR/Cas9-edited Pparγ. As applicable, the models were analyzed for levels of energy metabolism, glucose metabolism, and metabolic profile of extramedullary adipose tissue, as well as the osteocyte transcriptome, mitochondrial function, bioenergetics, insulin signaling, and oxidative stress. Circulating sclerostin levels of γOTKO male and female mice were not different from control mice. Male γOTKO mice exhibited a high energy phenotype characterized by increased respiration, heat production, locomotion and food intake. This high energy phenotype in males did not correlate with "beiging" of peripheral adipose depots. However, both sexes showed a trend for reduced fat mass and apparent insulin resistance without changes in glucose tolerance, which correlated with decreased osteocytic responsiveness to insulin measured by AKT activation. The transcriptome of osteocytes isolated from γOTKO males suggested profound changes in cellular metabolism, fuel transport, mitochondria dysfunction, insulin signaling and increased oxidative stress. In MLO-Y4 osteocytes, PPARG deficiency correlated with highly active mitochondria, increased ATP production, and accumulation of reactive oxygen species (ROS). PPARG in male osteocytes acts as a molecular break on mitochondrial function, and protection against oxidative stress and ROS accumulation. It also regulates osteocyte insulin signaling and fuel usage to produce energy. These data provide insight into the connection between osteocyte bioenergetics and their sex-specific contribution to the balance of systemic energy metabolism. These findings support the concept that the skeleton controls systemic energy expenditure via osteocyte metabolism. • Osteocytes regulate systemic energy metabolism via their bioenergetics. • PPARG protein acts as a "molecular break" of osteocyte mitochondrial activity. • PPARG deficiency activates TCA cycle, oxidative stress and ROS accumulation. • PPARG controls osteocyte insulin signaling and fuel utilization. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
10. Calebin A attenuated inflammation in RAW264.7 macrophages and adipose tissue to improve hepatic glucose metabolism and hyperglycemia in high-fat diet-fed obese mice.
- Author
-
Anwar, Choirul, Lin, Jing-Ru, Tsai, Mei-Ling, Ho, Chi-Tang, and Lai, Ching-Shu
- Subjects
- *
ADIPOSE tissues , *GLUCOSE metabolism , *ADIPOSE tissue physiology , *HYPERGLYCEMIA , *OBESITY , *INSULIN , *INFLAMMATION - Abstract
The increased incidence of obesity, which become a global health problem, requires more functional food products with minor side and excellent effects. Calebin A (CbA) is a non-curcuminoid compound, which is reported to be an effective treatment for lipid metabolism and thermogenesis. However, its ability and mechanism of action in improving obesity-associated hyperglycemia remain unclear. This study was designed to explore the effect and mechanism of CbA in hyperglycemia via improvement of inflammation and glucose metabolism in the adipose tissue and liver in high-fat diet (HFD)-fed mice. After 10 weeks fed HFD, obese mice supplemented with CbA (25 and 100 mg/kg) for another 10 weeks showed a remarkable reducing adiposity and blood glucose. CbA modulated M1/M2 macrophage polarization, ameliorated inflammatory cytokines, and restored adiponectin as well as Glut 4 expression in the adipose tissue. In the in vitro study, CbA attenuated pro-inflammatory markers while upregulated anti-inflammatory IL-10 in LPS + IFNγ-generated M1 phenotype macrophages. In the liver, CbA attenuated steatosis, inflammatory infiltration, and protein levels of inflammatory TNF-α and IL-6. Moreover, CbA markedly upregulated Adiponectin receptor 1, AMPK, and insulin downstream Akt signaling to improve glycogen content and increase Glut2 protein. These findings indicated that CbA may be a novel therapeutic approach to treat obesity and hyperglycemia phenotype targeting on adipose inflammation and hepatic insulin signaling. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Altered glucose metabolism in Alzheimer's disease: Role of mitochondrial dysfunction and oxidative stress.
- Author
-
Dewanjee, Saikat, Chakraborty, Pratik, Bhattacharya, Hiranmoy, Chacko, Leena, Singh, Birbal, Chaudhary, Anupama, Javvaji, Kalpana, Pradhan, Saumya Ranjan, Vallamkondu, Jayalakshmi, Dey, Abhijit, Kalra, Rajkumar Singh, Jha, Niraj Kumar, Jha, Saurabh Kumar, Reddy, P. Hemachandra, and Kandimalla, Ramesh
- Subjects
- *
ALZHEIMER'S disease , *GLUCOSE metabolism , *LYSOSOMES , *METABOLISM , *ADVANCED glycation end-products , *OXIDATIVE stress , *RECEPTOR for advanced glycation end products (RAGE) , *MITOCHONDRIA - Abstract
Increasing evidence suggests that abnormal cerebral glucose metabolism is largely present in Alzheimer's disease (AD). The brain utilizes glucose as its main energy source and a decline in its metabolism directly reflects on brain function. Weighing on recent evidence, here we systematically assessed the aberrant glucose metabolism associated with amyloid beta and phosphorylated tau accumulation in AD brain. Interlink between insulin signaling and AD highlighted the involvement of the IRS/PI3K/Akt/AMPK signaling, and GLUTs in the disease progression. While shedding light on the mitochondrial dysfunction in the defective glucose metabolism, we further assessed functional consequences of AGEs (advanced glycation end products) accumulation, polyol activation, and other contributing factors including terminal respiration, ROS (reactive oxygen species), mitochondrial permeability, PINK1/parkin defects, lysosome-mitochondrial crosstalk, and autophagy/mitophagy. Combined with the classic plaque and tangle pathologies, glucose hypometabolism with acquired insulin resistance and mitochondrial dysfunction potentiate these factors to exacerbate AD pathology. To this end, we further reviewed AD and DM (diabetes mellitus) crosstalk in disease progression. Taken together, the present work discusses the emerging role of altered glucose metabolism, contributing impact of insulin signaling, and mitochondrial dysfunction in the defective cerebral glucose utilization in AD. [Display omitted] • A decline in brain glucose levels and metabolism is pathologically linked to AD and dementia. • Insulin resistance and mitochondrial dysfunction are implicated in classic plaque and tangle pathologies of AD. • Aberrant IRS/PI3K/Akt/AMPK signaling and abnormal GLUT activities are directly linked to the etiology of neurodegeneration. • Defective glucose metabolism-induced polyol activation, AGEs accumulation and oxidative stress contribute to AD pathogenesis. • Abnormal glucose metabolism endorses cerebral autophagy/mitophagy dysregulation by hampering lysosome-mitochondrial functions. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
12. The INSR/AKT/mTOR pathway regulates the pace of myogenesis in a syndecan-3-dependent manner.
- Author
-
Jones, Fiona K., Phillips, Alexander M., Jones, Andrew R., and Pisconti, Addolorata
- Subjects
- *
MYOGENESIS , *INSULIN receptors , *STEM cells , *MUSCLE cells , *PROTEIN-tyrosine kinases , *MUSCLE regeneration - Abstract
• A phosphoproteomic survey of muscle stem cell progeny reveals that syndecans-3 regulates tyrosine kinase receptor-mediated signal transduction at multiple levels, including insulin/INSR signaling as one of the most enriched pathways. • The presence of syndecan-3 in muscle stem cells during the early stages of myogenesis inhibits insulin/INSR signaling via AKT/mTOR leading to muscle stem cell progeny expansion. • Syndecan-3 downregulation during the late stages of myogenesis releases the block on AKT/mTOR activation by insulin/INSR, leading to muscle stem cell progeny differentiation. • Syndecan-3 acts as a timekeeper of myogenesis, by functioning as a molecular switch for insulin signaling in muscle stem cell progeny. Muscle stem cells (MuSCs) are indispensable for muscle regeneration. A multitude of extracellular stimuli direct MuSC fate decisions from quiescent progenitors to differentiated myocytes. The activity of these signals is modulated by coreceptors such as syndecan-3 (SDC3). We investigated the global landscape of SDC3-mediated regulation of myogenesis using a phosphoproteomics approach which revealed, with the precision level of individual phosphosites, the large-scale extent of SDC3-mediated regulation of signal transduction in MuSCs. We then focused on INSR/AKT/mTOR as a key pathway regulated by SDC3 during myogenesis and mechanistically dissected SDC3-mediated inhibition of insulin receptor signaling in MuSCs. SDC3 interacts with INSR ultimately limiting signal transduction via AKT/mTOR. Both knockdown of INSR and inhibition of AKT restore Sdc3−/− MuSC differentiation to wild type levels. Since SDC3 is rapidly downregulated at the onset of differentiation, our study suggests that SDC3 acts a timekeeper to restrain proliferating MuSC response and prevent premature differentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
13. Cardiac substrate utilization in heart failure: Where is the relevance of SGLT2 inhibition?
- Author
-
Mann, Pascal Alexander and Lehrke, Michael
- Published
- 2022
- Full Text
- View/download PDF
14. TRAF6-mediated ubiquitination of AKT1 in the nucleus occurs in a β-arrestin2-dependent manner upon insulin stimulation.
- Author
-
Hu, Li, Liu, Haiping, Ma, Haixiang, Zeng, Xingyue, Cao, Yongkai, Liu, Bing, Li, Huijun, and Zhang, Xiaohan
- Subjects
- *
UBIQUITINATION , *INSULIN , *UBIQUITIN ligases , *PROTEIN kinase B , *NUCLEOCYTOPLASMIC interactions , *INSULIN receptors , *CELL anatomy - Abstract
[Display omitted] AKT, also known as protein kinase B (PKB), serves as a crucial regulator of numerous biological functions, including cell growth, metabolism, and tumorigenesis. Increasing evidence suggests that the kinase activity of AKT is regulated via ubiquitination by various E3 ligase enzymes in response to different stimuli. However, the molecular mechanisms underlying insulin-induced AKT ubiquitination are not yet fully understood. Here, we show that activation of the insulin receptor (IR) leads to enhanced ubiquitination of AKT1 at K8 and K14 residues, facilitated by the cytosolic E3 ubiquitin ligase enzyme, TRAF6. Further investigation using AKT1 mutants with modified nucleocytoplasmic shuttling properties reveals that TRAF6-mediated AKT1 ubiquitination occurs within the nucleus in a β-Arr2-dependent manner. The nuclear entry of TRAF6 depends on importin β1, while β-Arr2 regulates this process by facilitating the interaction between TRAF6 and importin β1. Additionally, the ubiquitination of AKT1 is essential for its translocation to the activated IR on the plasma membrane, where it plays a functional role in recruiting Glut4 and facilitating glucose uptake. This study uncovers the cellular components and processes involved in insulin-induced ubiquitination and activation of AKT1, providing insights and detailed strategies for manipulating AKT1. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Multi-step regulation of microRNA expression and secretion into small extracellular vesicles by insulin.
- Author
-
Lino, Marsel, Garcia-Martin, Ruben, Muñoz, Vitor Rosetto, Ruiz, Gabriel Palermo, Nawaz, Allah, Brandão, Bruna Brasil, Dreyfus, Jonathan, Pan, Hui, and Kahn, C. Ronald
- Abstract
Tissues release microRNAs (miRNAs) in small extracellular vesicles (sEVs) including exosomes, which can regulate gene expression in distal cells, thus acting as modulators of local and systemic metabolism. Here, we show that insulin regulates miRNA secretion into sEVs from 3T3-L1 adipocytes and that this process is differentially regulated from cellular expression. Thus, of the 53 miRNAs upregulated and 66 miRNAs downregulated by insulin in 3T3-L1 sEVs, only 12 were regulated in parallel in cells. Insulin regulated this process in part by phosphorylating hnRNPA1, causing it to bind to AU-rich motifs in miRNAs, mediating their secretion into sEVs. Importantly, 43% of insulin-regulated sEV-miRNAs are implicated in obesity and insulin resistance. These include let-7 and miR-103, which we show regulate insulin signaling in AML12 hepatocytes. Together, these findings demonstrate an important layer to insulin's regulation of adipose biology and provide a mechanism of tissue crosstalk in obesity and other hyperinsulinemic states. [Display omitted] • Insulin differentially regulates miRNA content in adipocytes and their sEVs • miRNA sorting depends on sequence motifs and RNA-binding protein phosphorylation • Insulin-regulated sEV miRNAs are involved in regulation of metabolic function • Adipocyte-derived miRNAs provide a novel mechanism of tissue crosstalk in obesity Adipose tissue is a major source of circulating exosomal miRNAs. Here, Lino et al. demonstrate that adipocyte secretion of miRNAs is regulated by insulin and this process involves the interaction of RNA-binding proteins, leading to the secretion of miRNAs enriched in those controlling metabolism in distant tissues, including hepatocytes. Thus, extracellular miRNAs are a new layer of endocrine regulation by insulin. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Methionine supplementation during a hydrogen peroxide challenge alters components of insulin signaling and antioxidant proteins in subcutaneous adipose explants from dairy cows.
- Author
-
Ma, N., Liang, Y., Coleman, D.N., Li, Y., Ding, H., Liu, F., Cardoso, F.F., Parys, C., Cardoso, F.C., Shen, X., and Loor, J.J.
- Subjects
- *
LIPOLYSIS , *GLUCOSE transporters , *FATTY acid synthases , *NF-kappa B , *HEPATOCYTE growth factor , *DAIRY cattle , *HYDROGEN peroxide , *INSULIN - Abstract
Enhanced postruminal supply of methionine (Met) during the peripartal period alters protein abundance of insulin, AA, and antioxidant signaling pathways in subcutaneous adipose tissue (SAT). Whether SAT is directly responsive to supply of Met and can induce molecular alterations is unknown. Our objective was to examine whether enhanced Met supply during an oxidative stress challenge in vitro alters insulin, AA, inflammation, and antioxidant signaling-related protein networks. Four late-lactation Holstein cows (average 27.0 kg of milk per day) were used for SAT collection. Tissue was incubated in duplicate for 4 h in a humidified incubator with 5% CO 2 at 37°C according to the following experimental design: control medium with an "ideal" profile of essential AA (CTR; Lys:Met 2.9:1), CTR plus 100 μ M H 2 O 2 (HP), or CTR with greater Met supply plus 100 μ M H 2 O 2 (HPMET; Lys:Met 2.5:1). Molecular targets associated with insulin signaling, lipolysis, antioxidant nuclear factor, erythroid 2 like 2 (NFE2L2), inflammation, and AA metabolism were determined through reverse-transcription quantitative PCR and western blotting. Data were analyzed using the MIXED procedure of SAS 9.4 (SAS Institute Inc.). Among proteins associated with insulin signaling, compared with CTR, HP led to lower abundance of phosphorylated AKT serine/threonine kinase (p-AKT) and solute carrier family 2 member 4 (SLC2A4; insulin-induced glucose transporter). Although incubation with HPMET restored abundance of SLC2A4 to levels in the CTR and upregulated abundance of fatty acid synthase (FASN) and phosphorylated 5′-prime-AMP-activated protein kinase (p-AMPK), it did not alter p-AKT, which remained similar to HP. Among proteins associated with AA signaling, compared with CTR, challenge with HP led to lower abundance of phosphorylated mechanistic target of rapamycin (p-MTOR), and HPMET did not restore abundance to CTR levels. Among inflammation-related targets studied, incubation with HPMET led to greater protein abundance of nuclear factor kappa B subunit p65 (NFKB-RELA). The response in NFKB observed with HPMET was associated with a marked upregulation of the antioxidant transcription regulator NFE2L2 and the antioxidant enzyme glutathione peroxidase 1 (GPX1). No effects of treatment were detected for mRNA abundance of proinflammatory cytokines or antioxidant enzymes, underscoring the importance of post-transcriptional regulation. Overall, data indicated that short-term challenge with H 2 O 2 was particularly effective in reducing insulin and AA signaling. Although a greater supply of Met had little effect on those pathways, it seemed to restore the protein abundance of the insulin-induced glucose transporter. Overall, the concomitant upregulation of key inflammation and antioxidant signaling proteins when a greater level of Met was supplemented to oxidant-challenged SAT highlighted the potential role of this AA in regulating the inflammatory response and oxidant status. Further studies should be conducted to assess the role of postruminal supply of Met and other AA in the regulation of immune, antioxidant, and metabolic systems in peripartal cow adipose tissue. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
17. Arsenic disrupts neuronal insulin signaling through increasing free PI3K-p85 and decreasing PI3K activity.
- Author
-
Wisessaowapak, Churaibhon, Watcharasit, Piyajit, and Satayavivad, Jutamaad
- Subjects
- *
INSULIN receptors , *PROTEIN-tyrosine kinases , *ARSENIC , *PHOSPHATIDYLINOSITOL 3-kinases , *INSULIN - Abstract
• Arsenic impaired neuronal insulin signaling a non-competitive like manner. • Arsenic decreased intrinsic tyrosine protein kinase activity of the insulin receptor. • Arsenic increased free PI3K-p85 and its interaction with IRS1. • Arsenic suppressed basal and insulin stimulated PI3K activity. Previously, we reported that prolonged arsenic exposure impaired neuronal insulin signaling. Here we have further identified novel molecular mechanisms underlying neuronal insulin signaling impairment by arsenic. Arsenic treatment altered insulin dose-response curve and reduced maximum insulin response in differentiated human neuroblastoma SH-SY5Y cells, suggesting that arsenic hindered neuronal insulin signaling in a non-competitive like manner. Mechanistically, arsenic suppressed insulin receptor (IR) kinase activity, as witnessed by a decreased insulin-activated autophosphorylation of IR at Y1150/1151. Arsenic decreased the level of insulin receptor substrate 1 (IRS1) but increased the protein ratio between PI3K regulatory subunit, p85, and PI3K catalytic subunit, p110. Interestingly, co-immunoprecipitation demonstrated that arsenic did not alter a level of PI3K-p110/PI3K-p85 complex while increased PI3K-p85 levels in a PI3K-p110 depletion supernatant resulted from PI3K-p110 immunoprecipitation. These results indicated that arsenic increased PI3K-p85 which was free from PI3K-p110 binding. In addition, arsenic significantly increased interaction between IRS1 and PI3K-p85 but not PI3K-p110, suggesting that there may be a fraction of free PI3K-p85 interacting with IRS1. In vitro PI3K activity demonstrated that arsenic lowered PI3K activity in both basal and insulin-stimulated conditions. These results suggested that the increase in free PI3K-p85 by arsenic might compete with PI3K heterodimer for the same IRS1 binding site, in turn blocking the activation of its catalytic subunit, PI3K-p110. Taken together, our results provide additional insights into mechanisms underlying the impairment of neuronal insulin signaling by arsenic through the reduction of IR autophosphorylation, the increase in free PI3K-p85, and the impeding of PI3K activity. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
18. Pyrazoles as novel protein tyrosine phosphatase 1B (PTP1B) inhibitors: An in vitro and in silico study.
- Author
-
Rocha, Sónia, Lucas, Mariana, Silva, Vera L.M., Gomes, Pedro M.O., Silva, Artur M.S., Araújo, Alberto N., Aniceto, Natália, Guedes, Rita C., Corvo, M. Luísa, Fernandes, Eduarda, and Freitas, Marisa
- Subjects
- *
PROTEIN-tyrosine phosphatase , *PHOSPHOPROTEIN phosphatases , *TYPE 2 diabetes , *PYRAZOLES , *INSULIN receptors , *MOLECULAR docking - Abstract
Type 2 diabetes mellitus (DM) is a complex chronic disorder and a major global health problem. Insulin resistance is the primary detectable abnormality and the main characteristic feature in individuals with type 2 DM. Protein tyrosine phosphatase 1B (PTP1B) is a key negative regulator of the insulin signaling pathway, which dephosphorylates insulin receptor and insulin receptor substrates, suppressing the insulin signaling cascade. Therefore, the inhibition of PTP1B has become a potential strategy in the management of type 2 DM. In this study, a library of 22 pyrazoles was evaluated here for the first time against human PTP1B activity, using a microanalysis screening system. The results showed that 5-(2-hydroxyphenyl)-3-{2-[3-(4-nitrophenyl)-1,2,3,4-tetrahydronaphthyl]}-1-phenylpyrazole 20 and 3-(2-hydroxyphenyl)-5-{2-[3-(4-methoxyphenyl)]naphthyl}pyrazole 22 excelled as the most potent inhibitors of PTP1B, through noncompetitive inhibition mechanism. These findings suggest that the presence of additional benzene rings as functional groups in the pyrazole moiety increases the ability of pyrazoles to inhibit PTP1B. The most active compounds showed selectivity over the homologous T-cell protein tyrosine phosphatase (TCPTP). Molecular docking analyses were performed and revealed a particular contact signature involving residues like TYR46, ASP48, PHE182, TYR46, ALA217 and ILE219. This study represents a significant beginning for the design of novel PTP1B inhibitors. [Display omitted] • Protein tyrosine phosphatase 1B (PTP1B) regulates the insulin signaling pathway. • PTP1B inhibition is currently a recognized promising target in type 2 diabetes. • Pyrazoles were shown to be effective inhibitors of PTP1B. • Molecular docking analysis revealed a particular contact signature of pyrazoles. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
19. PKCα: Prospects in Regulating Insulin Resistance and AD.
- Author
-
Mishra, Devanshi and Dey, Chinmoy Sankar
- Subjects
- *
INSULIN resistance , *PROTEIN kinase C , *ALZHEIMER'S disease , *CELLULAR signal transduction - Abstract
Protein kinase C alpha (PKCα) is known to participate in various signaling pathways due to its ubiquitous and dynamic characteristics. Previous studies report that PKCα abrogates peripheral insulin resistance, and recent publications show that it takes part in regulating Alzheimer's disease (AD). Based on evidence in the literature, we have highlighted how many of the substrates of PKCα in its signal transduction cascades are common in AD and diabetes and may have the capability to regulate both diseases simultaneously. Signaling pathways crosslinking these two diseases by PKCα have not been explored. Understanding the complexities of PKCα interactions with common molecules will deepen our understanding of its regulation of relevant pathophysiologies and, in the future, may broaden the possibility of using PKCα as a therapeutic target. Previous studies have indicated that protein kinase C alpha (PKCα) expression and activity is reduced in cancers, whereas its activity is enhanced in Alzheimer's disease (AD). Thus, an anticancer drug, which is supposed to increase PKCα levels in cancer, might create severe complications if the patient happens to be an AD patient or a prospective AD patient. Opposite expression profiles of PKCα occur in patients with insulin-resistant diabetes compared with insulin-sensitive subjects, opening up questions about the involvement of PKCα in diabetes. Neuronal insulin resistance is known to be associated with AD, and neuronal PKCα regulates many substrates, which seems to overlap between insulin-resistant diabetes and AD. Understanding this crosslinking of signaling molecules in both insulin-resistant diabetes and AD might lead to a better understanding of how PKCα regulates two dissimilar pathophysiologies. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
20. Elevated levels of alcohol dehydrogenase aggravate ethanol-evoked cardiac remodeling and contractile anomalies through FKBP5-yap-mediated regulation of ferroptosis and ER stress.
- Author
-
Lu, Qi, Qin, Xing, Chen, Chu, Yu, Wei, Lin, Jie, Liu, Xiaoyu, Guo, Rui, Reiter, Russel J., Ashrafizadeh, Milad, Yuan, Ming, and Ren, Jun
- Subjects
- *
ALCOHOL dehydrogenase , *CONTRACTILE proteins , *ETHANOL , *INSULIN receptors , *ACETALDEHYDE , *GLUCOSE intolerance , *CARRIER proteins , *YAP signaling proteins , *CELL death - Abstract
Alcohol intake provokes severe organ injuries including alcoholic cardiomyopathy with hallmarks of cardiac remodeling and contractile defects. This study examined the toxicity of facilitated ethanol metabolism in alcoholism-evoked changes in myocardial morphology and contractile function, insulin signaling and various cell death domains using cardiac-selective overexpression of alcohol dehydrogenase (ADH). WT and ADH mice were offered an alcohol liquid diet for 12 weeks prior to assessment of cardiac geometry, function, ER stress, apoptosis and ferroptosis. Alcohol intake provoked pronounced glucose intolerance, cardiac remodeling and contractile anomalies with apoptosis, ER stress, and ferroptosis, the effects were accentuated by ADH with the exception of global glucose intolerance. Hearts from alcohol ingesting mice displayed dampened insulin-stimulated phosphorylation of insulin receptor (tyr1146) and IRS-1 (tyrosine) along with elevated IRS-1 serine phosphorylation, the effect was augmented by ADH. Alcohol challenge dampened phosphorylation of Akt and GSK-3β, and increased phosphorylation of c-Jun and JNK, the effects were accentuated by ADH. Alcohol challenge promoted ER stress, FK506 binding protein 5 (FKBP5), YAP, apoptosis and ferroptosis, the effects were exaggerated by ADH. Using a short-term ethanol challenge model (3 g/kg, i.p., twice in three days), we found that inhibition of FKBP5-YAP signaling or facilitated ethanol detoxification by Alda-1 alleviated ethanol cardiotoxicity. In vitro study revealed that the ethanol metabolite acetaldehyde evoked cardiac contractile anomalies, lipid peroxidation, and apoptosis, the effects of which were mitigated by Alda-1, inhibition of ER stress, FKBP5 and YAP. These data suggest that facilitated ethanol metabolism via ADH exacerbates alcohol-evoked myocardial remodeling, functional defects, and insulin insensitivity possibly through a FKBP5-YAP-associated regulation of ER stress and ferroptosis. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. Silencing ANGPTL8 reduces mouse preadipocyte differentiation and insulin signaling.
- Author
-
Ghosh, Anindya, Leung, Yat Hei, Yu, Jeffrey, Sladek, Robert, Chénier, Isabelle, Oppong, Abel K., Peyot, Marie-Line, Madiraju, S.R. Murthy, Al-Khairi, Irina, Thanaraj, Thangavel Alphonse, Abubaker, Jehad, Al-Mulla, Fahd, Prentki, Marc, and Abu-Farha, Mohamed
- Subjects
- *
ADIPOGENESIS , *FAT cells , *INSULIN , *GENE expression , *LIPOPROTEIN lipase , *EXTRACELLULAR space , *ADIPOSE tissues - Abstract
ANGPTL8, expressed mainly in the liver and adipose tissue, regulates the activity of lipoprotein lipase (LPL) present in the extracellular space and triglyceride (TG) metabolism through its interaction with ANGPTL3 and ANGPTL4. Whether intracellular ANGPTL8 can also exert effects in tissues where it is expressed is uncertain. ANGPTL8 expression was low in preadipocytes and much increased during differentiation. To better understand the role of intracellular ANGPTL8 in adipocytes and assess whether it may play a role in adipocyte differentiation, we knocked down its expression in normal mouse subcutaneous preadipocytes. ANGPTL8 knockdown reduced adipocyte differentiation, cellular TG accumulation and also isoproterenol-stimulated lipolysis at day 7 of differentiation. RNA-Seq analysis of ANGPTL8 siRNA or control siRNA transfected SC preadipocytes on days 0, 2, 4 and 7 of differentiation showed that ANGPTL8 knockdown impeded the early (day 2) expression of adipogenic and insulin signaling genes, PPARγ, as well as genes related to extracellular matrix and NF-κB signaling. Insulin mediated Akt phosphorylation was reduced at an early stage during adipocyte differentiation. This study based on normal primary cells shows that ANGPTL8 has intracellular actions in addition to effects in the extracellular space, like modulating LPL activity. Preadipocyte ANGPTL8 expression modulates their differentiation possibly via changes in insulin signaling gene expression. • ANGPTL8 has intracellular actions besides its inhibitory effect on LPL activity. • ANGPTL8 knockdown in preadipocytes impairs their differentiation and adipogenesis. • ANGPTL8 modulates the expression of insulin and PPARγ signaling related genes. • ANGPTL8 controls the expression of genes related to NF-κB signaling and to ECM. • Preadipocyte ANGPTL8 modulates their differentiation by regulating insulin signaling. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. GT-11 impairs insulin signaling through modulation of sphingolipid metabolism in C2C12 myotubes.
- Author
-
Rustamov, Javokhir, Roh, Yoon-Seok, Hong, Jin Tae, and Yoo, Hwan-Soo
- Subjects
- *
AMIDASES , *INSULIN , *HIGH performance liquid chromatography , *INSULIN regulation , *GENE silencing , *METABOLISM - Abstract
Sphingolipids are involved in the regulation of insulin signaling, which is linked to the development of insulin resistance, leading to diabetes mellitus. We aimed to study whether modulation of sphingolipid levels by GT-11 may regulate insulin signaling in C2C12 myotubes. We investigated the effects of sphingolipid metabolism on Akt phosphorylation and glucose uptake using C2C12 myotubes. Either GT-11, an inhibitor of dihydroceramide desaturase 1 and S1P lyase, or siRNA targeting Sgpl1 , the gene encoding the enzyme, was employed to determine the effect of sphingolipid metabolism modulation on insulin signaling. Western blotting and glucose uptake assays were used to evaluate the effect of treatments on insulin signaling. Sphingolipid metabolites were analyzed by high performance liquid chromatography (HPLC). Treatment with GT-11 resulted in decreased Akt phosphorylation and reduced glucose uptake. Silencing the Sgpl1 gene, which encodes S1P lyase, mimicked these findings, suggesting the potential for regulating insulin signaling through S1P lyase modulation. GT-11 modulated sphingolipid metabolism, inducing the accumulation of sphingolipids. Using PF-543 and ARN14974 to inhibit sphingosine kinases and acid ceramidase, respectively, we identified a significant interplay between sphingosine, S1P lyase, and insulin signaling. Treatment with either exogenous sphingosine or palmitic acid inhibited Akt phosphorylation, and reduced S1P lyase activity. Our findings highlight the importance of close relationship between sphingolipid metabolism and insulin signaling in C2C12 myotubes, pointing to its potential therapeutic relevance for diabetes mellitus. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. Impacts of glutamate, an exercise-responsive metabolite on insulin signaling.
- Author
-
Xing, Xiaorui, Sun, Qin, Wang, Ruwen, Wang, Yibing, and Wang, Ru
- Subjects
- *
EXCITATORY amino acid antagonists , *INSULIN , *INSULIN sensitivity , *METABOLIC disorders , *LIPID metabolism , *GLUCOSE metabolism - Abstract
Disruption of the insulin signaling pathway leads to insulin resistance (IR). IR is characterized by impaired glucose and lipid metabolism. Elevated levels of circulating glutamate are correlated with metabolic indicators and may potentially predict the onset of metabolic diseases. Glutamate receptor antagonists have significantly enhanced insulin sensitivity, and improved glucose and lipid metabolism. Exercise is a well-known strategy to combat IR. The aims of our narrative review are to summarize preclinical and clinical findings to show the correlations between circulating glutamate levels, IR and metabolic diseases, discuss the causal role of excessive glutamate in IR and metabolic disturbance, and present an overview of the exercise-induced alteration in circulating glutamate levels. A literature search was conducted to identify studies on glutamate, insulin signaling, and exercise in the PubMed database. The search covered articles published from December 1955 to January 2024, using the search terms of "glutamate", "glutamic acid", "insulin signaling", "insulin resistance", "insulin sensitivity", "exercise", and "physical activity". Elevated levels of circulating glutamate are correlated with IR. Excessive glutamate can potentially hinder the insulin signaling pathway through various mechanisms, including the activation of ectopic lipid accumulation, inflammation, and endoplasmic reticulum stress. Glutamate can also modify mitochondrial function through Ca2+ and induce purine degradation mediated by AMP deaminase 2. Exercise has the potential to decrease circulating levels of glutamate, which can be attributed to accelerated glutamate catabolism and enhanced glutamate uptake. Glutamate may act as a mediator in the exercise-induced improvement of insulin sensitivity. [Display omitted] • Circulating levels of glutamate are correlated with the risk of developing IR and common metabolic diseases. • Dysregulation of the glutamate/receptor axis may play a causal role in the development of IR and metabolic disturbances. • Exercise has been shown to decrease the level of glutamate in circulation. • Glutamate may serve as an exercise-responsive metabolite that improves insulin sensitivity and metabolism. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. The many actions of insulin in skeletal muscle, the paramount tissue determining glycemia.
- Author
-
Sylow, Lykke, Tokarz, Victoria L., Richter, Erik A., and Klip, Amira
- Abstract
As the principal tissue for insulin-stimulated glucose disposal, skeletal muscle is a primary driver of whole-body glycemic control. Skeletal muscle also uniquely responds to muscle contraction or exercise with increased sensitivity to subsequent insulin stimulation. Insulin's dominating control of glucose metabolism is orchestrated by complex and highly regulated signaling cascades that elicit diverse and unique effects on skeletal muscle. We discuss the discoveries that have led to our current understanding of how insulin promotes glucose uptake in muscle. We also touch upon insulin access to muscle, and insulin signaling toward glycogen, lipid, and protein metabolism. We draw from human and rodent studies in vivo , isolated muscle preparations, and muscle cell cultures to home in on the molecular, biophysical, and structural elements mediating these responses. Finally, we offer some perspective on molecular defects that potentially underlie the failure of muscle to take up glucose efficiently during obesity and type 2 diabetes. Skeletal muscle plays a key role in glycemic control. In this Review, Sylow et al. discuss the scientific discoveries related to insulin stimulation of glucose uptake in skeletal muscle, including the development of insulin resistance in type 2 diabetes and the impact of exercise. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
25. The Inhibition of Metabolic Inflammation by EPA Is Associated with Enhanced Mitochondrial Fusion and Insulin Signaling in Human Primary Myotubes.
- Author
-
Sergi, Domenico, Luscombe-Marsh, Natalie, Heilbronn, Leonie K, Birch-Machin, Mark, Naumovski, Nenad, Lionetti, Lilla', Proud, Christopher G, Abeywardena, Mahinda Y, and O'Callaghan, Nathan
- Subjects
- *
EICOSAPENTAENOIC acid , *PEROXISOME proliferator-activated receptors , *MITOCHONDRIA , *INSULIN , *PALMITIC acid , *MEMBRANE potential , *MITOCHONDRIAL membranes , *GLUCOSE metabolism , *INFLAMMATION prevention , *RESEARCH , *SKELETAL muscle , *CELL culture , *INFLAMMATION , *BIOLOGICAL transport , *RESEARCH methodology , *MEDICAL cooperation , *EVALUATION research , *CELLULAR signal transduction , *COMPARATIVE studies , *DNA-binding proteins , *INSULIN resistance , *FATTY acids - Abstract
Background: Sustained fuel excess triggers low-grade inflammation that can drive mitochondrial dysfunction, a pivotal defect in the pathogenesis of insulin resistance in skeletal muscle.Objectives: This study aimed to investigate whether inflammation in skeletal muscle can be prevented by EPA, and if this is associated with an improvement in mitochondrial fusion, membrane potential, and insulin signaling.Methods: Human primary myotubes were treated for 24 h with palmitic acid (PA, 500 μM) under hyperglycemic conditions (13 mM glucose), which represents nutrient overload, and in the presence or absence of EPA (100 μM). After the treatments, the expression of peroxisome proliferator-activated receptor γ coactivator 1-α (PPARGC1A) and IL6 was assessed by q-PCR. Western blot was used to measure the abundance of the inhibitor of NF-κB (IKBA), mitofusin-2 (MFN2), mitochondrial electron transport chain complex proteins, and insulin-dependent AKT (Ser473) and AKT substrate 160 (AS 160; Thr642) phosphorylation. Mitochondrial dynamics and membrane potential were evaluated using immunocytochemistry and the JC-1 (tetraethylbenzimidazolylcarbocyanine iodide) dye, respectively. Data were analyzed using 1-factor ANOVA followed by Tukey post hoc test.Results: Nutrient excess activated the proinflammatory NFκB signaling marked by a decrease in IKBA (40%; P < 0.05) and the upregulation of IL6 mRNA (12-fold; P < 0.001). It also promoted mitochondrial fragmentation (53%; P < 0.001). All these effects were counteracted by EPA. Furthermore, nutrient overload-induced drop in mitochondrial membrane potential (6%; P < 0.05) was prevented by EPA. Finally, EPA inhibited fuel surplus-induced impairment in insulin-mediated phosphorylation of AKT (235%; P < 0.01) and AS160 (49%; P < 0.05).Conclusions: EPA inhibited NFκB signaling, which was associated with an attenuation of the deleterious effects of PA and hyperglycemia on both mitochondrial health and insulin signaling in human primary myotubes. Thus, EPA might preserve skeletal muscle metabolic health during sustained fuel excess but this requires confirmation in human clinical trials. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
26. Influence of obese phenotype on metabolic profile, inflammatory mediators and stemness of hADSC in adipose tissue.
- Author
-
Rawal, Komal, Patel, Tushar P., Purohit, Kishan M., Israni, Kashish, Kataria, Vyakhaya, Bhatt, Hiren, and Gupta, Sarita
- Abstract
Unhealthy dietary practices, sedentary life style and lack of physical exercise in developing countries like India are major contributors of metabolic syndrome like obesity and diabetes. Obesity in Indians is defined at Body Mass Index (BMI, kg/m
2 ) >25 and characterized as metabolically obese. A preliminary study performed to explore ramification of obesity on metabolic profile of adipose tissue and adipose derived stem cells (ADSC) from control and obese Indians. Adipose tissue/lipoaspirates from both control (BMI ≤ 23) subjects, and non-diabetic obese Indians subjects (BMI ≥ 25), were scrutinized for expressions of lipogenic genes, inflammatory mediators, stored adipokine levels, and insulin signaling proteins. Further, hADSC were isolated and immune-phenotyped from both the subject groups. Comparative assessments between chADSC and ohADSC were carried out for growth kinetics, expressions of pluripotent genes, adipogenic transcriptional factors, RUNX2, inflammatory mediators (IM), insulin signaling proteins, adipogenic and osteogenic differentiation. Adipose tissue of obese subjects depicted high leptin and resistin levels with reduced adiponectin levels. Expressions of IM and insulin signaling proteins were elevated compared to those of control subjects. hADSC of obese subjects demonstrated diminished proliferation, altered pluripotent genes, aggravated inflammation, adipogenesis with reduced osteogenesis. hADSC of obese had established insulin resistance compared to those of control subjects. This is the first study that describes hADSC of metabolically obese Indians have insulin resistance at lower BMI compared to Caucasians exemplifying plausible role in diminishing stemness of hADSC. Study alarms Indians to restore healthy dietary habits and assess quality of hADSC in regenerative therapy. • First comparative study on hADSC in metabolically obese Indians. • hADSC of obese committed to adipogenic lineage, reduced osteogenesis. • hADSC of obese Indians have insulin resistance, reduced MYF5, 1st population study. • High levels of inflammatory markers in hADSC and adipose tissue of obese. • Altered stored adipokines and lipogenic genes of adipose tissue in Indian obese. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
27. Protective effect of the hydroalcoholic extract from Lampaya medicinalis Phil. (Verbenaceae) on palmitic acid- impaired insulin signaling in 3T3-L1 adipocytes.
- Author
-
Ormazabal, Paulina, Herrera, Karin, Cifuentes, Mariana, Paredes, Adrián, Morales, Glauco, and Cruz, Gonzalo
- Subjects
PHYTOTHERAPY ,BLOOD sugar ,BLOOD sugar monitoring ,CELLULAR signal transduction ,FAT cells ,FATTY acids ,GLUCOSE ,INSULIN ,INSULIN resistance ,PHOSPHORYLATION ,STATISTICAL sampling ,WESTERN immunoblotting ,PLANT extracts ,RANDOMIZED controlled trials ,CYTOTOXINS - Abstract
• The range of 0.01–10.0 μg/mL of HEL is not cytotoxic in 3T3-L1 adipocytes. • HEL restores PA-disrupted phosphorylation of IRS-1/Akt/AS160 in 3T3-L1 adipocytes. • HEL ameliorates PA-impaired glucose uptake in 3T3-L1 adipocytes. Obesity is strongly associated with insulin resistance (IR). IR at the molecular level may be defined as a diminished activation of insulin signaling-related molecules (IRS-1/Akt/AS160) as well as reduced glucose uptake. Subject with obesity have elevated plasma levels of saturated fatty acids, such as palmitic acid (PA), which triggers insulin signaling disruption in vivo and in vitro. Infusions of Lampaya medicinalis Phil. (Verbenaceae) are used in folk medicine of Northern Chile to counteract inflammatory diseases. Hydroethanolic extracts of lampaya (HEL) contain considerable amounts of flavonoids that may explain the biological activity of the plant. The aim of this study was to assess whether HEL exposure protects against PA-disrupted insulin signaling and glucose uptake in adipocytes. Cytotoxicity of a range of HEL concentrations (0.01–10 μg/mL) was evaluated in 3T3-L1 adipocytes. Cells were exposed or not to 0.1 μg/mL of HEL before adding 0.65 mM PA or vehicle and incubated with 100 nM insulin (or vehicle) for 15 min. Phosphorylation of Tyr-IRS-1, Ser-Akt, Thr-AS160 was evaluated by Western blot. Glucose uptake was assessed using the 2-NBDG analogue. HEL was not cytotoxic at any concentration assessed. PA-induced reduction in insulin-stimulated phosphorylation of IRS-1, Akt and AS160 and glucose uptake were abolished by co-treatment with HEL. These findings give new insights about the effect of HEL ameliorating PA- impaired IRS-1/Akt/AS160 pathway and glucose uptake in adipocytes. More studies should focus on lampaya, since might represent a preventive approach in individuals whose circulating PA levels contribute to IR. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
28. Modulation of redox and insulin signaling underlie the anti-hyperglycemic and antioxidant effects of diphenyl diselenide in zebrafish.
- Author
-
dos Santos, Matheus M., de Macedo, Gabriel T., Prestes, Alessandro S., Ecker, Assis, Müller, Talise E., Leitemperger, Jossiele, Fontana, Bárbara D., Ardisson-Araújo, Daniel M.P., Rosemberg, Denis B., and Barbosa, Nilda V.
- Subjects
- *
DIPHENYL diselenide , *ORGANOSELENIUM compounds , *GLUCOSE transporters , *INSULIN , *BLOOD sugar , *INSULIN receptors , *FRUCTOOLIGOSACCHARIDES - Abstract
The organic selenium compound diphenyl diselenide (DD) has been recognized as an antioxidant and neuroprotective agent, exerting an anti-hyperglycemic effect in experimental models of diabetes. However, the precise mechanisms involved in the protection are unclear. Using the zebrafish (Danio rerio) as a model organism, here we investigated biomarkers underlying the protective effects of DD against hyperglycemia, targeting in a transcriptional approach the redox and insulin-signaling pathway. Fish were fed on a diet containing DD (3 mg/kg) for 74 days. In the last 14 days, they were exposed to a 111 mM glucose solution to induce a hyperglycemic state. DD reduced blood glucose levels as well as normalized the brain mRNA transcription of four insulin receptors-coding genes (Insra1, Insra2, Insrb1, Insrb2), which were down-regulated by glucose. DD alone caused an up-regulation of relative mRNA transcription in both Insra receptors and glucose transporter 3 genes. DD counteracted hyperglycemia-induced lipid peroxidation, protein and thiol depletion. Along with the decreased activity of antioxidant enzymes SOD and GPx, the brain of hyperglycemic fish presented a reduction in mRNA transcription of FoxO3A , FoxO3B, Nrf2, GPx3A, SOD1 , and SOD2 genes. Besides normalizing the transcriptional levels, DD caused an up-regulation of relative mRNAs that encode Nrf2, FoxO1A, FOXO3A , GPx4A , PTP1B, AKT and SelP. Collectively, our findings suggest that the antioxidant and anti-hyperglycemic actions of DD in a zebrafish diabetes model are likely associated with the regulation of the oxidative stress resistance and the insulin-signaling pathway and that could be related to the modulation at mRNA level of two important transcription factors, Nrf2 and FoxO. Image 1 • Hyperglycemia caused oxidative damage and reduced the antioxidant defenses in brain. • Hyperglycemia down-regulated brain transcription of insulin receptors-coding genes. • Hyperglycemia decreased the brain mRNA transcription of Nrf2 and FoxO -coding genes. • DD counteracted all effects hyperglycemia-induced in redox and insulin signaling. • DD up-regulated mRNAs that encode Insra, Glut, Nrf2, FoxO, GPx, PTP1B, AKT and SelP. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
29. Metabolic Communication and Healthy Aging: Where Should We Focus Our Energy?
- Author
-
Smith, Hannah J., Sharma, Arpit, and Mair, William B.
- Subjects
- *
CELLULAR aging , *LONGEVITY , *AGING , *VESICLES (Cytology) - Abstract
Aging is associated with a loss of metabolic homeostasis and plasticity, which is causally linked to multiple age-onset pathologies. The majority of the interventions—genetic, dietary, and pharmacological—that have been found to slow aging and protect against age-related disease in various organisms do so by targeting central metabolic pathways. However, targeting metabolic pathways chronically and ubiquitously makes it difficult to define the downstream effects responsible for lifespan extension and often results in negative effects on growth and health, limiting therapeutic potential. Insight into how metabolic signals are relayed between tissues, cells, and organelles opens up new avenues to target metabolic regulators locally rather than globally for healthy aging. In this review, we discuss the pro-longevity effects of targeting metabolic pathways in specific tissues and how these interventions communicate with distal cells to modulate aging. These studies may be crucial in designing interventions that promote longevity without negative health consequences. Multiple metabolic pathways can be manipulated to slow the rate of aging, but doing so ubiquitously can have negative physiological consequences. Smith et al. review evidence that targeting central age-related metabolic pathways in specific tissues can promote healthy aging and might circumnavigate detrimental effects. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
30. Dietary Fat and Sugar Differentially Affect β-Adrenergic Stimulation of Cardiac ERK and AKT Pathways in C57BL/6 Male Mice Subjected to High-Calorie Feeding.
- Author
-
Ashraf, Sadia, Yilmaz, Gizem, Chen, Xu, and Harmancey, Romain
- Subjects
- *
THREONINE , *SUCROSE , *WESTERN diet , *GLUCOSE tolerance tests , *HIGH-fat diet , *CARBOHYDRATE content of food , *FAT , *BODY composition , *FAT content of food , *ANIMAL experimentation , *BLOOD sugar , *INGESTION , *CELLULAR signal transduction , *INSULIN , *TRANSFERASES , *GENES , *LIPIDS , *MICE - Abstract
Background: High dietary fat and sugar promote cardiac hypertrophy independently from an increase in blood pressure. The respective contribution that each macronutrient exerts on cardiac growth signaling pathways remains unclear.Objective: The goal of this study was to investigate the mechanisms by which high amounts of dietary fat and sugar affect cardiac growth regulatory pathways.Methods: Male C57BL/6 mice (9 wk old; n = 20/group) were fed a standard rodent diet (STD; kcal% protein-fat-carbohydrate, 29-17-54), a high-fat diet (HFD; 20-60-20), a high-fat and high-sugar Western diet (WD; 20-45-35), a high-sugar diet with mixed carbohydrates (HCD; 20-10-70), or a high-sucrose diet (HSD; 20-10-70). Body composition was assessed weekly by EchoMRI. Whole-body glucose utilization was assessed with an intraperitoneal glucose tolerance test. After 6 wk on diets, mice were treated with saline or 20 mg/kg isoproterenol (ISO), and the activity of cardiac growth regulatory pathways was analyzed by immunoblotting. Data were analyzed by ANOVA with data from the STD group included for references only.Results: Compared with HCD and HSD, WD and HFD increased body fat mass 2.7- to 3.8-fold (P < 0.001), induced glucose intolerance (P < 0.001), and increased insulin concentrations >1.5-fold (P < 0.05), thereby enhancing basal and ISO-stimulated AKT phosphorylation at both threonine 308 and serine 473 residues (+25-63%; P < 0.05). Compared with HFD, the high-sugar diets potentiated ISO-mediated stimulation of the glucose-sensitive kinases PYK2 (>47%; P < 0.05 for HCD and HSD) and ERK (>34%; P < 0.05 for WD, HCD, and HSD), thereby leading to increased phosphorylation of protein synthesis regulator S6K1 at threonine 389 residue (>64%; P < 0.05 for WD, HCD, and HSD).Conclusions: Dietary fat and sugar affect cardiac growth signaling pathways in C57BL/6 mice through distinct and additive mechanisms. The findings may provide new insights into the role of overnutrition in pathological cardiac remodeling. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
31. Diabetes as a risk factor for Alzheimer's disease in the Middle East and its shared pathological mediators.
- Author
-
Jayaraj, Richard L., Azimullah, Sheikh, and Beiram, Rami
- Abstract
The incidence of Alzheimer's disease (AD) has risen exponentially worldwide over the past decade. A growing body of research indicates that AD is linked to diabetes mellitus (DM) and suggests that impaired insulin signaling acts as a crucial risk factor in determining the progression of this devastating disease. Many studies suggest people with diabetes, especially type 2 diabetes, are at higher risk of eventually developing Alzheimer's dementia or other dementias. Despite nationwide efforts to increase awareness, the prevalence of Diabetes Mellitus (DM) has risen significantly in the Middle East and North African (MENA) region which might be due to rapid urbanization, lifestyle changes, lack of physical activity and rise in obesity. Growing body of evidence indicates that DM and AD are linked because both conditions involve impaired glucose homeostasis and altered brain function. Current theories and hypothesis clearly implicate that defective insulin signaling in the brain contributes to synaptic dysfunction and cognitive deficits in AD. In the periphery, low-grade chronic inflammation leads to insulin resistance followed by tissue deterioration. Thus insulin resistance acts as a bridge between DM and AD. There is pressing need to understand on how DM increases the risk of AD as well as the underlying mechanisms, due to the projected increase in age related disorders. Here we aim to review the incidence of AD and DM in the Middle East and the possible link between insulin signaling and ApoE carrier status on Aβ aggregation, tau hyperphosphorylation, inflammation, oxidative stress and mitochondrial dysfunction in AD. We also critically reviewed mutation studies in Arab population which might influence DM induced AD. In addition, recent clinical trials and animal studies conducted to evaluate the efficiency of anti-diabetic drugs have been reviewed. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
32. Leucine increases mitochondrial metabolism and lipid content without altering insulin signaling in myotubes.
- Author
-
Rivera, Madison E., Lyon, Emily S., Johnson, Michele A., and Vaughan, Roger A.
- Subjects
- *
LIPID metabolism , *LEUCINE , *INSULIN , *INSULIN resistance , *INSULIN receptors , *MYOBLASTS - Abstract
Elevated circulating branched-chain amino acids (BCAA) such as leucine have been consistently correlated with increasing severity of insulin resistance across numerous populations. BCAA may promote insulin resistance through either mTOR-mediated suppression of insulin receptor substrate-1 or through the accumulation of toxic BCAA catabolites. Although the link between circulating BCAA and insulin resistance has been consistent, it has yet to be concluded if BCAA causally contribute to the development or worsening of insulin resistance. This work investigated the effect of leucine both with and without varying levels of insulin resistance on metabolism, metabolic gene expression, and insulin signaling. C2C12 myotubes were treated with and without varied concentrations of leucine up to 2 mM for 24 h both with and without varied levels of insulin resistance. Gene and protein expression were measured via qRT-PCR and Western blot, respectively. Mitochondrial metabolism was measured via O 2 consumption. Leucine at 2 mM increased oxidative metabolism as well as gene expression of mitochondrial biogenesis, which was associated with increased cellular lipid content. Despite increased lipid content of leucine-treated cells, neither acute nor chronic leucine treatment at 2 mM affected insulin signaling in insulin sensitive, mildly insulin resistant, or severely insulin resistant cells. Similarly, leucine at lower concentrations (0.25 mM, 0.5 mM, and 1 mM) did not alter insulin signaling either, regardless of insulin resistance. Leucine appears to improve myotube oxidative metabolism and related metabolic gene expression. And despite increased lipid content of leucine-treated cells, leucine does not appear to alter insulin sensitivity either acutely or chronically, regardless of level of insulin resistance. • Leucine upregulated myotube mitochondrial biogenic gene expression. • Leucine increased lipid content and mitochondrial metabolism of myotubes. • Leucine did not alter insulin signaling regardless of insulin sensitivity. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
33. Insulin signaling and insulin response in subcutaneous and retroperitoneal adipose tissue in Holstein cows during the periparturient period.
- Author
-
Kenéz, Á., Ruda, L., Dänicke, S., and Huber, K.
- Subjects
- *
ADIPOSE tissue physiology , *ADIPOSE tissues , *PARTURITION , *INSULIN receptors , *COWS , *SERUM , *PROTEIN expression , *INSULIN - Abstract
Adipose tissue response to endocrine stimuli, such as insulin, is crucial for metabolic adaptation at the onset of lactation in dairy cows. However, the exact molecular mechanisms behind this response are not well understood. Thus, the aim of this study was to determine the dynamics in protein expression and phosphorylation of key components in insulin signaling in subcutaneous (SCAT) and retroperitoneal (RPAT) adipose tissues of Holstein dairy cows. Furthermore, by ex vivo examinations, response to insulin was assessed in SCAT and RPAT at different time points during the periparturient period. Biopsy samples were taken 42 d prepartum, and 1, 21, and 100 d postpartum. Insulin and glucose concentrations were measured in blood serum in consecutive serum samples from d −42 until d +100. After parturition, the majority of the key components were downregulated in both adipose tissues but recovered by d +100. The extent of hormone-sensitive lipase phosphorylation increased postpartum and remained high throughout the experimental period. Strong differences in molecular response were observed between the 2 depots. The RPAT expressed a remarkably greater extent of AMP-activated kinase phosphorylation compared with SCAT, indicating that AMP-activated kinase as an energy sensor is highly active particularly in RPAT in times of energy scarcity. Consequently, this depot expressed a greater extent of hormone-sensitive lipase phosphorylation over the whole experimental period. Insulin response after parturition appeared to be greater in RPAT too, due to the significantly greater expression of the insulin receptor at d +21 and +100. Although insulin concentrations in plasma were low postpartum, the depot-specific changes in molecular modulation of insulin signaling and insulin response suggested that both adipose tissue depots studied were contributing to the periparturient homeorhetic adaptation, although most likely to a different extent. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
34. Protocatechuic acid exhibits hepatoprotective, vasculoprotective, antioxidant and insulin-like effects in dexamethasone-induced insulin-resistant rats.
- Author
-
El-Sonbaty, Yomna A., Suddek, Ghada M., Megahed, Nirmeen, and Gameil, Nariman M.
- Subjects
- *
ASPARTATE aminotransferase , *NITRIC-oxide synthases , *GLUCOSE intolerance , *NADPH oxidase , *INSULIN resistance , *SOLEUS muscle , *HYPERGLYCEMIA - Abstract
Protocatechuic acid (PCA), the natural phenolic antioxidant, reportedly exhibited hypoglycemic and insulin-like effects. Recent studies have reported its cardioprotective effect in glucocorticoid (GC)-induced hypertensive rats. Nevertheless, its beneficial role has not been investigated in the setting of GCs excess-induced insulin resistance. This study aimed to investigate the possible protective potential and the plausible mechanisms of pretreatment with PCA against GCs-induced insulin resistance, liver steatosis and vascular dysfunction. Insulin resistance was induced in male Wistar rats by a 7-day treatment with dexamethasone (DEX) (1 mg/kg/day, i.p.). PCA (50, 100 mg/kg/day, orally) was started 7 days before DEX administration and continued during the test period. PCA significantly and dose-dependently attenuated DEX-induced a) glucose intolerance (↓ AUC OGTT), b) hyperglycemia (↓ fasting blood glucose), c) impaired insulin sensitivity [↓fasting plasma insulin and homeostasis model assessment of insulin resistance (HOMA-IR) index)] and d) dyslipidemia (↓total cholesterol, triglycerides, low-density lipoprotein-cholesterol and very low-density lipoprotein-cholesterol). PCA mitigated DEX-induced liver steatosis with associated reduction in serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activity. Moreover, PCA ameliorated DEX-induced vascular dysfunction and enhanced ACh-induced relaxation in aortic rings. The metabolic ameliorating effects of PCA might be attributed to the enhanced insulin signaling in soleus muscles (↑AKT phosphorylation) and mitigating gluconeogenesis (↓ hepatic mRNA expression of phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6Pase). The vasculoprotective effect of PCA might be related to its ability to restore normal mRNA expression of [endothelial nitric oxide synthase (eNOS) and NADPH Oxidase 4 (NOX4)]. PCA restored normal oxidative balance [↓ oxidant species, malondialdehyde (MDA) and (↑ antioxidant superoxide dismutase (SOD)]. The findings herein reveal for the first time that PCA may be taken as a supplement with GCs to limit their metabolic and vascular side effects through its hypoglycemic, insulin-sensitizing, hypolipidemic and antioxidant effects. Image 1 • PCA enhanced insulin-mediated AKT phosphorylation in DEX-treated rats. • PCA attenuated DEX-induced upregulation in the expression of PEPCK and G6Pase. • PCA normalized the expression of eNOS and NOX 4. • PCA restored the oxidant/antioxidant balance. • PCA exerted hypolipidemic effect and ameliorated liver steatosis. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
35. Insulin and adipokine signaling and their cross-regulation in postmortem human brain.
- Author
-
Wang, Hoau-Yan, Capuano, Ana W., Khan, Amber, Pei, Zhe, Lee, Kuo-Chieh, Bennett, David A., Ahima, Rexford S., Arnold, Steven E., and Arvanitakis, Zoe
- Subjects
- *
LEPTIN , *ADIPOKINES , *INSULIN , *LEPTIN receptors , *TYPE 2 diabetes , *INSULIN receptors , *ALZHEIMER'S disease - Abstract
Aberrant insulin and adipokine signaling has been implicated in cognitive decline associated with both type 2 diabetes mellitus and neurodegenerative diseases. We established methods that reliably measure insulin, adiponectin and leptin signaling, and their crosstalk, in thawed postmortem mid-frontal cortical tissue from cognitively normal older subjects with a short postmortem interval. Insulin-evoked insulin receptor (IR) activation increases activated, tyrosine-phosphorylated IRβ on tyrosine residues 960, 1150, and 1151, insulin receptor substrate-1 recruitment to IRβ and phosphorylated RAC-α-serine/threonine-protein kinase. Adiponectin augments, but leptin inhibits, insulin signaling. Adiponectin activates adiponectin receptors to induce APPL1 binding to adiponectin receptor 1 and 2 and T-cadherin and downstream adenosine monophosphate–dependent protein kinase phosphorylation. Insulin inhibited adiponectin-induced signaling. In addition, leptin-induced leptin receptor (OB-R) signaling promotes Janus kinase 2 recruitment to OB-R and Janus kinase 2 and downstream signal transducer and activator of transcription 3 phosphorylation. Insulin enhanced leptin signaling. These data demonstrate insulin and adipokine signaling interactions in human brain. Future studies can use these methods to examine insulin, adiponectin, and leptin metabolic dysregulation in aging and disease states, such as type 2 diabetes and Alzheimer's disease–related dementias. • Insulin, adipokine signaling, and their crosstalk can be measured in postmortem human brain. • Adipokine enhances but leptin inhibits insulin signaling. • Insulin reduces adiponectin but potentiates leptin signaling. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
36. Programmed increases in LXRα induced by paternal alcohol use enhance offspring metabolic adaptation to high-fat diet induced obesity.
- Author
-
Chang, Richard C., Thomas, Kara N., Bedi, Yudhishtar S., and Golding, Michael C.
- Abstract
Paternally inherited alterations in epigenetic programming are emerging as relevant factors in numerous disease states, including the growth and metabolic defects observed in fetal alcohol spectrum disorders. In rodents, chronic paternal alcohol use induces fetal growth restriction, as well as sex-specific alterations in insulin signaling and lipid homeostasis in the offspring. Based on previous studies, we hypothesized that the observed metabolic irregularities are the consequence of paternally inherited alterations liver x receptor (LXR) activity. Male offspring of alcohol-exposed sires were challenged with a high-fat diet and the molecular pathways controlling glucose and lipid homeostasis assayed for LXR-induced alterations. Similar to findings in studies employing LXR agonists we found that the male offspring of alcohol-exposed sires display resistance to diet-induced obesity and improved glucose homeostasis when challenged with a high-fat diet. This improved metabolic adaptation is mediated by LXRα trans-repression of inflammatory cytokines, releasing IKKβ inhibition of the insulin signaling pathway. Interestingly, paternally programmed increases in LXRα expression are liver-specific and do not manifest in the pancreas or visceral fat. These studies identify LXRα as a key mediator of the long-term metabolic alterations induced by preconception paternal alcohol use. • Chronic paternal alcohol use induces up-regulation of LXRα in the male offspring. • Male offspring of alcohol-exposed fathers are protected from diet-induced obesity. • Paternally-inherited up-regulation of LXRα only manifests in the liver. • Improved metabolic adaptation is linked to LXRα suppression of cytokine production. • Male offspring exhibit the same phenotypes observed in studies of LXR agonists. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
37. Insulin signaling in insulin-dysregulated Icelandic horses.
- Author
-
Frers, F., Delarocque, J., Feige, K., Huber, K., and Warnken, T.
- Subjects
- *
INSULIN receptors , *PROTEIN kinase B , *INSULIN , *HORSES , *ADENOSINE monophosphate , *WESTERN immunoblotting , *PROTEIN kinases - Abstract
• Insulin signaling was effective in insulin dysregulated (ID) horses. • ID horses had a higher extent of phosphorylation of the insulin receptor in liver tissue. • ID horses had higher baseline mTOR phosphorylation compared to insulin-sensitive horses. • Insulin dysregulation appeared to be associated with alterations of mTOR signaling. The underlying molecular mechanisms leading to insulin dysregulation are poorly understood in horses. Therefore, this study aimed to determine if insulin dysregulation is associated with an altered basal expression and extent of phosphorylation of key proteins of the insulin signaling cascade in liver (LT), muscle (MT), and subcutaneous adipose tissue (AT) under basal and stimulated conditions. Twelve Icelandic horses were subjected (1) to an oral glucose (Gluc PO) challenge and (2) to an intravenous (Ins IV) insulin challenge in a crossover study. Biopsies of LT, MT, and AT were taken in vivo under basal conditions and after Gluc PO and Ins IV stimulation. Corresponding insulin levels were measured by an equine optimized ELISA (Mercodia AB, Uppsala). Insulin levels ≥ 110 µIU/mL at 120 min indicated that six horses were insulin dysregulated (HI), while six were not (NI). Gluc PO stimulation resulted in a more pronounced hyperinsulinemia and hyperglycemia in HI horses compared to NI horses. Western blot analysis of key proteins of the insulin signaling cascade revealed an enhanced phosphorylation of the insulin receptor (InsR) under Gluc PO (P = 0.001) and Ins IV stimulation (P = 0.017) within LT, but not in MT and AT. Phosphorylation of protein kinase B was enhanced under Gluc PO stimulation in all tissues and under Ins IV stimulation in MT and AT, while phosphorylation of adenosine monophosphate protein kinase α was reduced after glucose administration (P = 0.005) in all horses. Interestingly, HI horses had significantly higher amounts of phosphorylated mechanistic target of rapamycin (mTOR) in MT (P = 0.049), irrespective of any stimulation. In LT, the amount of phosphorylated mTOR decreased under Gluc PO conditions in HI horses, while an increase was observed in NI horses (P = 0.015). A major limitation was the inclusion of only Icelandic horses of advanced age since insulin dysregulation could be related to both the equine metabolic syndrome and/or pituitary pars intermedia dysfunction. In summary, insulin signaling appeared to be maintained in both HI and NI Icelandic horses, although post-receptor alterations were observed. Thus, ID might be an equine-specific metabolic condition, in which alterations of the mTOR signaling pathway may play a crucial role, as emphasized by higher mTOR phosphorylation in HI horses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. Protein tyrosine phosphatase 1B is a regulator of alpha-actinin4 in the glomerular podocyte.
- Author
-
Hsu, Ming-Fo, Ito, Yoshihiro, Singh, Jai Prakash, Hsu, Shu-Fang, Wells, Alan, Jen, Kuang-Yu, Meng, Tzu-Ching, and Haj, Fawaz G.
- Subjects
- *
PHOSPHOPROTEIN phosphatases , *PROTEIN-tyrosine phosphatase , *PROTEIN crosslinking , *MASS spectrometry , *KIDNEY physiology , *TYROSINE , *CYTOSKELETAL proteins , *INSULIN receptors - Abstract
Glomerular podocytes are instrumental for the barrier function of the kidney, and podocyte injury contributes to proteinuria and the deterioration of renal function. Protein tyrosine phosphatase 1B (PTP1B) is an established metabolic regulator, and the inactivation of this phosphatase mitigates podocyte injury. However, there is a paucity of data regarding the substrates that mediate PTP1B actions in podocytes. This study aims to uncover novel substrates of PTP1B in podocytes and validate a leading candidate. To this end, using substrate-trapping and mass spectroscopy, we identified putative substrates of this phosphatase and investigated the actin cross-linking cytoskeletal protein alpha-actinin4. PTP1B and alpha-actinin4 co-localized in murine and human glomeruli and transiently transfected E11 podocyte cells. Additionally, podocyte PTP1B deficiency in vivo and culture was associated with elevated tyrosine phosphorylation of alpha-actinin4. Conversely, reconstitution of the knockdown cells with PTP1B attenuated alpha-actinin4 tyrosine phosphorylation. We demonstrated co-association between alpha-actinin4 and the PTP1B substrate-trapping mutant, which was enhanced upon insulin stimulation and disrupted by vanadate, consistent with an enzyme-substrate interaction. Moreover, we identified alpha-actinin4 tandem tyrosine residues 486/487 as mediators of its interaction with PTP1B. Furthermore, knockdown studies in E11 cells suggest that PTP1B and alpha-actinin4 are modulators of podocyte motility. These observations indicate that PTP1B and alpha-actinin4 are likely interacting partners in a signaling node that modulates podocyte function. Targeting PTP1B and plausibly this one of its substrates may represent a new therapeutic approach for podocyte injury that warrants additional investigation. [Display omitted] • PTP1B and alpha-actinin4 partially co-localize in the murine and human glomeruli. • Alpha-actinin4 tyrosine phosphorylation is elevated upon PTP1B deficiency. • PTP1B substrate trapping mutant co-associates with alpha-actinin4 upon insulin stimulation. • Alpha-actinin4 tandem tyrosine residues 486/487 mediate the interaction with PTP1B. • PTP1B and alpha-actinin4 are likely modulators of podocyte motility. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
39. Piperine modulates IR/Akt/GLUT4 pathways to mitigate insulin resistance: Evidence from animal and computational studies.
- Author
-
Prasad, Monisha, Jayaraman, Selvaraj, Natarajan, Sathan Raj, Veeraraghavan, Vishnu Priya, Krishnamoorthy, Rajapandiyan, Gatasheh, Mansour K., Palanisamy, Chella Perumal, and Elrobh, Mohamed
- Subjects
- *
INSULIN resistance , *INSULIN sensitivity , *HIGH-fat diet , *METABOLIC disorders , *BIOACTIVE compounds , *INSULIN - Abstract
The global prevalence of diabetes mellitus is rising, especially in India. Medicinal herbs, whether used alone or in combination with conventional medicines, have shown promise in managing diabetes and improving overall well-being. Piperine (PIP), a major bioactive compound found in pepper, is gaining attention for its beneficial properties. This study aimed to assess whether PIP could alleviate diabetes by targeting insulin pathway-related molecules in the adipose tissue of rats on a high-fat diet (HFD). After 60 days on the HFD, rats received PIP at a dose of 40 mg/kg body weight for one month. The results showed that PIP significantly improved metabolic indicators, antioxidant enzymes, and carbohydrate metabolic enzymes. It also regulated the mRNA and protein expression of insulin signaling, which had been disrupted by the diet and sucrose intake. Molecular docking analysis also revealed strong binding of PIP to key diabetes-related regulatory proteins, including Akt (−6.2 kcal/mol), IR (−7.02 kcal/mol), IRS-1 (−6.86 kcal/mol), GLUT4 (−6.24 kcal/mol), AS160 (−6.28 kcal/mol), and β-arrestin (−6.01 kcal/mol). Hence, PIP may influence the regulation of glucose metabolism through effective interactions with these proteins, thereby controlling blood sugar levels due to its potent antilipidemic and antioxidant properties. In conclusion, our study provides in vivo experimental evidence against the HFD-induced T2DM model for the first time, making PIP a potential natural remedy to enhance the quality of life for diabetic patients and aid in their management. • PIP enhances insulin and glucose tolerance in rats on HFD. • PIP increases IR/Akt/GLUT4 signaling, boosting insulin sensitivity and glucose uptake. • Computational data indicate PIP interacts with IR/Akt/GLUT4 signaling molecules, potentially amplifying their activity. • PIP holds potential as a natural remedy for IR and associated metabolic disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
40. Sympathetic overdrive and unrestrained adipose lipolysis drive alcohol-induced hepatic steatosis in rodents.
- Author
-
Zhou, Chunxue, Ruiz, Henry H., Ling, Li, Maurizi, Giulia, Sakamoto, Kenichi, Liberini, Claudia G., Wang, Ling, Stanley, Adrien, Egritag, Hale E., Sanz, Sofia M., Lindtner, Claudia, Butera, Mary A., and Buettner, Christoph
- Abstract
Hepatic steatosis is a key initiating event in the pathogenesis of alcohol-associated liver disease (ALD), the most detrimental organ damage resulting from alcohol use disorder. However, the mechanisms by which alcohol induces steatosis remain incompletely understood. We have previously found that alcohol binging impairs brain insulin action, resulting in increased adipose tissue lipolysis by unrestraining sympathetic nervous system (SNS) outflow. Here, we examined whether an impaired brain–SNS–adipose tissue axis drives hepatic steatosis through unrestrained adipose tissue lipolysis and increased lipid flux to the liver. We examined the role of lipolysis, and the brain–SNS–adipose tissue axis and stress in alcohol induced hepatic triglyceride accumulation in a series of rodent models: pharmacological inhibition of the negative regulator of insulin signaling protein-tyrosine phosphatase 1β (PTP1b) in the rat brain, tyrosine hydroxylase (TH) knockout mice as a pharmacogenetic model of sympathectomy, adipocyte specific adipose triglyceride lipase (ATGL) knockout mice, wildtype (WT) mice treated with β3 adrenergic agonist or undergoing restraint stress. Intracerebral administration of a PTP1b inhibitor, inhibition of adipose tissue lipolysis and reduction of sympathetic outflow ameliorated alcohol induced steatosis. Conversely, induction of adipose tissue lipolysis through β3 adrenergic agonism or by restraint stress worsened alcohol induced steatosis. Brain insulin resistance through upregulation of PTP1b, increased sympathetic activity, and unrestrained adipose tissue lipolysis are key drivers of alcoholic steatosis. Targeting these drivers of steatosis may provide effective therapeutic strategies to ameliorate ALD. • Alcohol binging induced fatty liver is reduced by CNS inhibition of the negative regulator of insulin signaling PTP1b. • Reduction of adipose tissue lipolysis prevents alcohol induced hepatic steatosis. • Reducing sympathetic outflow improves alcohol induced fatty liver. • Stress and β3 adrenergic agonism synergize with alcohol in increasing lipolysis and driving hepatic triglyceride accumulation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
41. Salts and energy balance: A special role for dietary salts in metabolic syndrome.
- Author
-
Brey, Christopher W., Akbari-Alavijeh, Safoura, Ling, Jun, Sheagley, Jordan, Shaikh, Bilal, Al-Mohanna, Futwan, Wang, Yi, Gaugler, Randy, and Hashmi, Sarwar
- Abstract
Dietary salts sodium (Na
+ ), potassium (K+ ), magnesium (Mg2+ ), and calcium (Ca2+ ) are important in metabolic diseases. Yet, we do not have sufficient understanding on the salts global molecular network in these diseases. In this systematic review we have pooled information to identify the general effect of salts on obesity, insulin resistance and hypertension. To assess the roles of salts in metabolic disorders by focusing on their individual effect and the network effect among these salts. We searched articles in PubMed, EMBASE and Google Scholar. We selected original laboratory research, systematic reviews, clinical trials, observational studies and epidemiological data that focused on dietary salts and followed the preferred reporting items for systematic review in designing the present systematic review. From the initial search of 2898 studies we selected a total of 199 articles that met our inclusion criteria and data extraction. Alterations in metabolic pathways associated with the sensitivity of sodium, potassium, magnesium and calcium may lead to obesity, hypertension, and insulin resistance. We found that the results of most laboratory research, animal studies and clinical trials are coherent but some research outcome are either inconsistent or inconclusive. Important of salts in metabolic disorder is evident. In order to assess the effects of dietary salts in metablic diseases, environmental factors, dietary habits, physical activity, and the microbiome, should be considered in any study. Although interest in this area of research continues to grow, the challenge is to integrate the action of these salts in metabolic syndrom. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
42. Dysregulation of PP2A-Akt interaction contributes to Sucrose non-fermenting related kinase (SNRK) deficiency induced insulin resistance in adipose tissue.
- Author
-
Li, Jie, An, Ran, Lai, Shuiqing, Li, Linlin, Liu, Simin, and Xu, Haiyan
- Abstract
We previously identified Sucrose non-fermenting related kinase (SNRK) as a regulator of adipose inflammation and energy homeostasis. In this study, we aimed to investigate the role of SNRK in insulin signaling in white (WAT) and brown adipose tissue (BAT). Adipose tissue specific (SNRK deficiency in both WAT and BAT) and BAT specific knockout mouse models were employed. Phosphoproteomic studies were conducted to identify the novel SNRK pathway regulating insulin signaling in adipose tissue. SNRK ablation is sufficient to inhibit insulin-stimulated AKT phosphorylation and glucose uptake in both WAT and BAT. Phosphoproteomic study using SNRK deficient versus wild type BAT samples revealed 99% reduction of phosphorylation on Serine 80 of PPP2R5D, the regulatory subunit of Protein phosphatase 2A (PP2A). Drastic (142.5-fold) induction of phosphorylation on Serine 80 of PPP2R5D was observed in SNRK-deficient primary brown adipocytes overexpressing SNRK compared to control protein. In vitro phosphorylation reaction followed by targeted phosphoproteomic detection further confirms that human recombinant SNRK is able to phosphorylate human recombinant PPP2R5D. Dephosphorylated PPP2R5D promotes constitutive assembly of PP2A-AKT complex, therefore inhibits insulin-induced AKT phosphorylation and subsequent glucose uptake in both BAT and WAT. Knockdown of PPP2R5D in adipocytes can improve insulin sensitivity in adipocytes without SNRK expression. Our findings demonstrate that SNRK regulates insulin signaling through controlling PPP2R5D phosphorylation, which subsequently impacts PP2A activity and then AKT phosphorylation in both WAT and BAT. SNRK may represent a promising potential target for treating insulin resistance-related metabolic disorders. • SNRK is essential for insulin-stimulated AKT phosphorylation in adipose tissue. • SNRK ablation causes insulin resistance in both white and brown adipose tissue. • Phosphoproteomic studies identify PPP2R5D as a novel substrate of SNRK. • SNRK regulates PP2A-AKT interaction through PPP2R5D phosphorylation. • Enhanced PP2A activity by SNRK ablation inhibits AKT phosphorylation. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
43. The role of skeletal muscle Akt in the regulation of muscle mass and glucose homeostasis.
- Author
-
Jaiswal, N., Gavin, M.G., Quinn III, W.J., Luongo, T.S., Gelfer, R.G., Baur, J.A., and Titchenell, P.M.
- Abstract
Skeletal muscle insulin signaling is a major determinant of muscle growth and glucose homeostasis. Protein kinase B/Akt plays a prominent role in mediating many of the metabolic effects of insulin. Mice and humans harboring systemic loss-of-function mutations in Akt2 , the most abundant Akt isoform in metabolic tissues, are glucose intolerant and insulin resistant. Since the skeletal muscle accounts for a significant amount of postprandial glucose disposal, a popular hypothesis in the diabetes field suggests that a reduction in Akt, specifically in skeletal muscle, leads to systemic glucose intolerance and insulin resistance. Despite this common belief, the specific role of skeletal muscle Akt in muscle growth and insulin sensitivity remains undefined. We generated multiple mouse models of skeletal muscle Akt deficiency to evaluate the role of muscle Akt signaling in vivo. The effects of these genetic perturbations on muscle mass, glucose homeostasis and insulin sensitivity were assessed using both in vivo and ex vivo assays. Surprisingly, mice lacking Akt2 alone in skeletal muscle displayed normal skeletal muscle insulin signaling, glucose tolerance, and insulin sensitivity despite a dramatic reduction in phosphorylated Akt. In contrast, deletion of both Akt isoforms (M-AktDKO) prevented downstream signaling and resulted in muscle atrophy. Despite the absence of Akt signaling, in vivo and ex vivo insulin-stimulated glucose uptake were normal in M-AktDKO mice. Similar effects on insulin sensitivity were observed in mice with prolonged deletion (4 weeks) of both skeletal muscle Akt isoforms selectively in adulthood. Conversely, short term deletion (2 weeks) of skeletal muscle specific Akt in adult muscles impaired insulin tolerance paralleling the effect observed by acute pharmacological inhibition of Akt in vitro. Mechanistically, chronic ablation of Akt induced mitochondrial dysfunction and activation of AMPK, which was required for insulin-stimulated glucose uptake in the absence of Akt. Together, these data indicate that chronic reduction in Akt activity alone in skeletal muscle is not sufficient to induce insulin resistance or prevent glucose uptake in all conditions. Therefore, since insulin-stimulated glucose disposal in skeletal muscle is markedly impaired in insulin-resistant states, we hypothesize that alterations in signaling molecules in addition to skeletal muscle Akt are necessary to perturb glucose tolerance and insulin sensitivity in vivo. • Deletion of skeletal muscle Akt2 alone does not reduce downstream insulin signaling or alter glucose homeostasis. • Inhibition of both skeletal muscle Akt isoforms prevents downstream signaling and results in muscle atrophy. • Chronic ablation of Akt in skeletal muscle does not block insulin-stimulated glucose uptake in vivo. • Prolonged Akt deficiency activates AMPK, which is required for insulin-stimulated glucose uptake in muscle lacking Akt. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
44. Xenobiotic mediated diabetogenesis: Developmental exposure to dichlorvos or atrazine leads to type 1 or type 2 diabetes in Drosophila.
- Author
-
Gupta, Himanshu Pawankumar, Jha, Rakesh Roshan, Ahmad, Humaira, Patel, Devendra Kumar, and Ravi Ram, Kristipati
- Subjects
- *
ATRAZINE , *TYPE 1 diabetes , *TYPE 2 diabetes , *ORGANOPHOSPHORUS pesticides , *DROSOPHILA , *XENOBIOTICS - Abstract
The increased incidence of diabetes to the magnitude of a global epidemic is attributed to non-traditional risk factors, including exposure to environmental chemicals. However, the contribution of xenobiotic exposure during the development of an organism to the etiology of diabetes is not fully addressed. Developing stages are more susceptible to chemical insult, but knowledge on the consequence of the same to the onset of diabetes is residual. In this context, by using Drosophila melanogaster having conserved Insulin/Insulin growth factor-like signaling (IIS) as well as glucose homeostasis as a model, we evaluated the potential of developmental exposure to dichlorvos (DDVP, an organophosphorus pesticide) or atrazine (herbicide) to cause diabetes in exposed organisms. Flies exposed to DDVP during their development display insulin deficiency or type 1 diabetes (T1D) while those exposed to atrazine show insulin resistance or type 2 diabetes (T2D), suggesting that exposure to these xenobiotics during organismal development can result in diabetes and that different mechanisms underlie pesticide mediated diabetes. We show that oxidative stress-mediated c-Jun N-terminal kinase (JNK) signaling activation underlies insulin resistance in flies exposed to atrazine during their development while DDVP-mediated T1D involves activation of caspase-mediated cell death pathway. Mitigation of oxidative stress through over-expression of SOD2 in atrazine (20μg/ml) exposed flies, revealed significantly decreased oxidative stress levels and reduced phosphorylation of JNK. Moreover, glucose and Akt phosphorylation levels in SOD2 over-expression flies exposed to atrazine were comparable to those in controls, suggesting restoration in insulin sensitivity. Therefore, exposure to xenobiotics during development is a common risk factor for the development of type 1 or type 2 diabetes. Accordingly, the present study cautions against the use of such diabetogenic pesticides. Also, mitigation of oxidative stress or anti-oxidant supplementation could be a potential therapy for xenobiotic mediated type 2 diabetes. Image 1 • Dichlorvos exposure during development leads to type 1 diabetes in Drosophila. • Activation of cell death pathway underlies DDVP mediated Type 1 diabetes. • Atrazine exposure during development causes type 2 diabetes in Drosophila. • Elevated oxidative stress along with insulin resistance in Atrazine exposed flies. • Oxidative stress alleviation in Atrazine exposed flies restores glucose homeostasis. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
45. Replacing Part of Glucose with Galactose in the Postweaning Diet Protects Female But Not Male Mice from High-Fat Diet-Induced Adiposity in Later Life.
- Author
-
Bouwman, Lianne M S, Fernández-Calleja, José M S, van der Stelt, Inge, Oosting, Annemarie, Keijer, Jaap, van Schothorst, Evert M, van der Stelt, Inge, and van Schothorst, Evert M
- Subjects
- *
MESODERM , *GALACTOSE , *WHITE adipose tissue , *OBESITY , *GLUCOSE , *INSULIN receptors - Abstract
Background: Duration of breastfeeding is positively associated with decreased adiposity and increased metabolic health in later life, which might be related to galactose.Objective: The aim of this study was to investigate if partial replacement of glucose with galactose in the postweaning diet had a metabolic programming effect.Methods: Male and female mice (C57BL/6JRccHsd) received an isocaloric diet (16 energy% fat; 64 energy% carbohydrates; 20 energy% protein) with either glucose (32 energy%) (GLU) or glucose + galactose (GLU + GAL, 16 energy% each) for 3 wk postweaning. Afterwards, all mice were switched to the same 40 energy% high-fat diet (HFD) for 9 wk to evaluate potential programming effects in an obesogenic environment. Data were analyzed within sex.Results: Female body weight (-14%) and fat mass (-47%) were significantly lower at the end of the HFD period (both P < 0.001) among those fed GLU + GAL than among those fed GLU; effects in males were in line with these findings but nonsignificant. Food intake was affected in GLU + GAL-fed females (+8% on postweaning diet, -9% on HFD) compared with GLU-fed females, but not for hypothalamic transcript levels at endpoint. Also, in GLU + GAL-fed females, serum insulin concentrations (-48%, P < 0.05) and the associated homeostasis model assessment of insulin resistance (HOMA-IR) were significantly lower ( P < 0.05) at endpoint, but there were no changes in pancreas morphology. In GLU + GAL-fed females, expression of insulin receptor substrate 2 (Irs2) (-27%, P < 0.01 ; -44%, P < 0.001) and the adipocyte size markers leptin (Lep) (-40%, P < 0.05; -63% , P < 0.05) and mesoderm-specific transcript homolog protein (Mest) (-80%, P < 0.05; -72%, P < 0.05) was lower in gonadal and subcutaneous white adipose tissue (WAT), respectively. Expression of insulin receptor substrate1 (Irs1) (-24%, P < 0.05) was only lower in subcutaneous WAT in GLU + GAL-fed females.Conclusions: Partial replacement of glucose with galactose, resulting in a 1:1 ratio mimicking lactose, in a 3-wk postweaning diet lowered body weight, adiposity, HOMA-IR, and expression of WAT insulin signaling in HFD-challenged female mice in later life. This suggests that prolonged galactose intake may improve metabolic and overall health in later life. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
46. Bombyxin/Akt signaling in relation to the embryonic diapause process of the silkworm, Bombyx mori.
- Author
-
Gu, Shi-Hong, Lin, Pei-Ling, and Hsieh, Hsiao-Yen
- Subjects
- *
SILKWORMS , *GLYCOGEN synthase kinase , *OVIPARITY - Abstract
• LY294002 inhibited phosphorylation of GSK-3β and Akt in Bombyx eggs. • Changes in Akt phosphorylation were different between diapause and developing eggs. • Changes in bombyxin-Z1 gene expression coincided with Akt phosphorylation. • Bombyxin/Akt signaling is related to Bombyx embryonic diapause process. Our previous study showed that phosphorylation of glycogen synthase kinase (GSK)-3β is related to the embryonic diapause process in Bombyx. However, the upstream signaling pathway was not clearly understood. In the present study, we examined bombyxin/Akt signaling in relation to the embryonic diapause process of B. mori. Results showed that GSK-3β phosphorylation stimulated by dechorionation was blocked by LY294002, a specific phosphatidylinositol 3-kinase (PI3K) inhibitor, indicating involvement of PI3K in GSK-3β phosphorylation in dechorionated eggs. Direct determination of Akt phosphorylation showed that dechorionation stimulated Akt phosphorylation. The Akt phosphorylation was blocked by LY294002. Temporal changes in Akt phosphorylation showed that different changing patterns exist between diapause and developing eggs. Relatively higher phosphorylation levels of Akt were detected between days 3 and 5 after oviposition in non-diapause eggs compared to those at the same stages in diapause eggs. Upon treatment with HCl, which prevents diapause initiation, Akt phosphorylation levels exhibited a later and much broader peak compared to diapause eggs. Examination of expression levels of the bombyxin-Z1 gene showed that in diapause eggs, a major peak occurred 1 day after oviposition, and its level then sharply decreased on day 2. However, in both non-diapause and HCl-treated eggs, a major broad peak was detected between days 1 and 4 after oviposition. These temporal changes in bombyxin-Z1 gene expression levels during embryonic stages coincided with changes in Akt phosphorylation, indicating that bombyxin-Z1 is likely an upstream signaling component for Akt phosphorylation. Taken together, our results indicated that PI3K/Akt is an upstream signaling pathway for GSK-3β phosphorylation and is associated with the diapause process of B. mori eggs. To our knowledge, this is the first study to demonstrate the potential correlation between bombyxin/Akt signaling and the embryonic diapause process. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
47. Reduced insulin action in muscle of high fat diet rats over the diurnal cycle is not associated with defective insulin signaling.
- Author
-
Small, Lewin, Brandon, Amanda E., Parker, Benjamin L., Deshpande, Vinita, Samsudeen, Azrah F., Kowalski, Greg M., Reznick, Jane, Wilks, Donna L., Preston, Elaine, Bruce, Clinton R., James, David E., Turner, Nigel, and Cooney, Gregory J.
- Abstract
Energy metabolism and insulin action follow a diurnal rhythm. It is therefore important that investigations into dysregulation of these pathways are relevant to the physiology of this diurnal rhythm. We examined glucose uptake, markers of insulin action, and the phosphorylation of insulin signaling intermediates in muscle of chow and high fat, high sucrose (HFHS) diet-fed rats over the normal diurnal cycle. HFHS animals displayed hyperinsulinemia but had reduced systemic glucose disposal and lower muscle glucose uptake during the feeding period. Analysis of gene expression, enzyme activity, protein abundance and phosphorylation revealed a clear diurnal regulation of substrate oxidation pathways with no difference in Akt signaling in muscle. Transfection of a constitutively active Akt2 into the muscle of HFHS rats did not rescue diet-induced reductions in insulin-stimulated glucose uptake. These studies suggest that reduced glucose uptake in muscle during the diurnal cycle induced by short-term HFHS-feeding is not the result of reduced insulin signaling. • Investigating metabolism in rodents over the diurnal cycle more accurately models normal animal physiology. • Diurnal regulation of substrate oxidation is altered in muscle of HFHS-fed rats. • There is a disconnect between glucose uptake and canonical insulin signaling in muscle. • Activation of Akt2 does not rescue diet-induced reductions in insulin-stimulated glucose uptake in muscle. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
48. Inositol pyrophosphates and Akt/PKB: Is the pancreatic β-cell the exception to the rule?
- Author
-
Kim, Jaeyoon, Darè, Elisabetta, Rajasekaran, Subu Surendran, Ryu, Sung Ho, Berggren, Per-Olof, and Barker, Christopher J.
- Subjects
- *
INOSITOL , *PYROPHOSPHATES , *INSULIN receptors , *THIAMIN pyrophosphate , *HIGH-fat diet , *PANCREATIC beta cells - Abstract
The inositol pyrophosphate, diphosphoinositol pentakisphosphate (IP 7), is thought to negatively regulate the critical insulin signaling protein Akt/PKB. Knockdown of the IP 7 -generating inositol hexakisphosphate kinase 1 (IP6K1) results in a concomitant increase in signaling through Akt/PKB in most cell types so far examined. Total in vivo knockout of IP6K1 is associated with a phenotype resistant to high-fat diet, due to enhanced Akt/PKB signaling in classic insulin regulated tissues, counteracting insulin resistance. In contrast, we have shown an important positive role for IP6K1 in insulin exocytosis in the pancreatic β-cell. These cells also possess functional insulin receptors and the feedback loop following insulin secretion is a key aspect of their normal function. Thus we examined the effect of silencing IP6K1 on the activation of Akt/PKB in β-cells. Silencing reduced the glucose-stimulated increase in Akt/PKB phosphorylation on T308 and S473. These effects were reproduced with the selective pan-IP6K inhibitor TNP. The likely explanation for IP 7 reduction decreasing rather than increasing Akt/PKB phosphorylation is that IP 7 is responsible for generating the insulin signal, which is the main source of Akt/PKB activation. In agreement, insulin receptor activation was compromised in TNP treated cells. To test whether the mechanism of IP 7 inhibition of Akt/PKB still exists in β-cells, we treated them at basal glucose with an insulin concentration equivalent to that reached during glucose stimulation. TNP potentiated the Akt/PKB phosphorylation of T308 induced by exogenous insulin. Thus, the IP 7 regulation of β-cell Akt/PKB is determined by two opposing forces, direct inhibition of Akt/PKB versus indirect stimulation via secreted insulin. The latter mechanism is dominant, masking the inhibitory effect. Consequently, pharmacological strategies to knock down IP6K activity might not have the same positive output in the β-cell as in other insulin regulated tissues. Unlabelled Image • Silencing IP6K1 inhibits glucose-induced Akt/PKB activation in pancreatic β-cells. • A pan IP6K inhibitor, TNP, also reduces glucose-stimulated Akt/PKB phosphorylation. • IP6K1 inhibition impairs Akt/PKB by curtailing insulin secretion and its autocrine feedback. • There are both positive and negative actions of IP 7 on Akt/PKB. • The dominant positive action of IP 7 on Akt/PKB is due to unique insulin autocrine feedback. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
49. Reduced biliverdin reductase-A levels are associated with early alterations of insulin signaling in obesity.
- Author
-
Cimini, Flavia Agata, Arena, Andrea, Barchetta, Ilaria, Tramutola, Antonella, Ceccarelli, Valentina, Lanzillotta, Chiara, Fontana, Mario, Bertoccini, Laura, Leonetti, Frida, Capoccia, Danila, Silecchia, Gianfranco, Di Cristofano, Claudio, Chiappetta, Caterina, Di Domenico, Fabio, Baroni, Marco Giorgio, Perluigi, Marzia, Cavallo, Maria Gisella, and Barone, Eugenio
- Subjects
- *
SERINE/THREONINE kinases , *INSULIN , *OBESITY , *FATTY liver , *INSULIN receptors , *INSULIN regulation - Abstract
Biliverdin reductase-A (BVR-A) is a serine/threonine/tyrosine kinase involved in the regulation of insulin signaling. In vitro studies have demonstrated that BVR-A is a substrate of the insulin receptor and regulates IRS1 by avoiding its aberrant activation, and in animal model of obesity the loss of hepatic BVR-A has been associated with glucose/insulin alterations and fatty liver disease. However, no studies exist in humans. Here, we evaluated BVR-A expression levels and activation in peripheral blood mononuclear cells (PBMC) from obese subjects and matched lean controls and we investigated the related molecular alterations of the insulin along with clinical correlates. We showed that BVR-A levels are significantly reduced in obese subjects and associated with a hyper-activation of the IR/IRS1/Akt/GSK-3β/AS160/GLUT4 pathway. Low BVR-A levels also associate with the presence of obesity, metabolic syndrome, NASH and visceral adipose tissue inflammation. These data suggest that the reduction of BVR-A may be responsible for early alterations of the insulin signaling pathway in obesity and in this context may represent a novel molecular target to be investigated for the comprehension of the process of insulin resistance development in obesity. • Reduced BVR-A protein levels were found in PBMC from obese subjects. • Low BVR-A are associated with reduced IRS1 inhibition. • Reduced IRS1 inhibition leads to insulin signaling hyper-activation. • Insulin signaling hyper-activation preserves euglycemia in obese subjects. • Insulin signaling hyper-activation precedes insulin resistance. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
50. Antipsychotics differentially regulate insulin, energy sensing, and inflammation pathways in hypothalamic rat neurons.
- Author
-
Kowalchuk, Chantel, Kanagasundaram, Pruntha, Belsham, Denise D., and Hahn, Margaret K.
- Subjects
- *
APPETITE stimulants , *INSULIN , *ANTIPSYCHOTIC agents , *NEURONS , *PSYCHIATRIC treatment , *ARIPIPRAZOLE - Abstract
• Antipsychotic (AP)s directly impact insulin and inflammatory pathways in hypothalamic neurons. • The MAPK pathway was consistently upregulated by all APs. • Only clozapine and aripiprazole inhibited pAKT and increased pAMPK. • Olanzapine and aripiprazole upregulated inflammatory pathways and BDNF. • APs have differential effects in the hypothalamus which do not align with clinical metabolic risk. Second generation antipsychotic (AP)s remain the gold-standard treatment for schizophrenia and are widely used on- and off-label for other psychiatric illnesses. However, these agents cause serious metabolic side-effects. The hypothalamus is the primary brain region responsible for whole body energy regulation, and disruptions in energy sensing (e.g. insulin signaling) and inflammation in this brain region have been implicated in the development of insulin resistance and obesity. To elucidate mechanisms by which APs may be causing metabolic dysregulation, we explored whether these agents can directly impact energy sensing and inflammation in hypothalamic neurons. The rat hypothalamic neuronal cell line, rHypoE-19, was treated with olanzapine (0.25–100 uM), clozapine (2.5–100 uM) or aripiprazole (5–20 uM). Western blots measured the energy sensing protein AMPK, components of the insulin signaling pathway (AKT, GSK3β), and components of the MAPK pathway (ERK1/2, JNK, p38). Quantitative real-time PCR was performed to determine changes in the mRNA expression of interleukin (IL)-6, IL-10 and brain derived neurotrophic factor (BDNF). Olanzapine (100 uM) and clozapine (100, 20 uM) significantly increased pERK1/2 and pJNK protein expression, while aripiprazole (20 uM) only increased pJNK. Clozapine (100 uM) and aripiprazole (5 and 20 uM) significantly increased AMPK phosphorylation (an orexigenic energy sensor), and inhibited insulin-induced phosphorylation of AKT. Olanzapine (100 uM) treatment caused a significant increase in IL-6 while aripiprazole (20 uM) significantly decreased IL-10. Olanzapine (100 uM) and aripiprazole (20 uM) increased BDNF expression. We demonstrate that antipsychotics can directly regulate insulin, energy sensing, and inflammatory pathways in hypothalamic neurons. Increased MAPK activation by all antipsychotics, alongside olanzapine-associated increases in IL-6 , and aripiprazole-associated decreases in IL-10 , suggests induction of pro-inflammatory pathways. Clozapine and aripiprazole inhibition of insulin-stimulated pAKT and increases in AMPK phosphorylation (an orexigenic energy sensor) suggests impaired insulin action and energy sensing. Conversely, olanzapine and aripiprazole increased BDNF , which would be expected to be metabolically beneficial. Overall, our findings suggest differential effects of antipsychotics on hypothalamic neuroinflammation and energy sensing. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.