1,667 results on '"high‐fat"'
Search Results
2. Maintenance of response to ketogenic diet therapy for drug-resistant epilepsy post diet discontinuation: A multi-centre case note review
- Author
-
Schoeler, Natasha E, Ridout, Deborah, Neal, Elizabeth G, Becirovic, Marisa, Whiteley, Victoria J, Meskell, Rachel, Lightfoot, Kathryn, Mills, Nicole, Ives, Tammy, Bara, Vanessa, Cameron, Emma, Thomas, Phillipa, Wilford, Ellen, Fox, Rachel, Fabe, Jennifer, Leong, Jia Yi, and Tan-Smith, Charlene
- Published
- 2024
- Full Text
- View/download PDF
3. Calorie restriction increases insulin sensitivity to promote beta cell homeostasis and longevity in mice
- Author
-
dos Santos, Cristiane, Cambraia, Amanda, Shrestha, Shristi, Cutler, Melanie, Cottam, Matthew, Perkins, Guy, Lev-Ram, Varda, Roy, Birbickram, Acree, Christopher, Kim, Keun-Young, Deerinck, Thomas, Dean, Danielle, Cartailler, Jean Philippe, MacDonald, Patrick E, Hetzer, Martin, Ellisman, Mark, and Arrojo e Drigo, Rafael
- Subjects
Medical Biochemistry and Metabolomics ,Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Diabetes ,Genetics ,Aging ,Nutrition ,1.1 Normal biological development and functioning ,Metabolic and endocrine ,Animals ,Caloric Restriction ,Insulin-Secreting Cells ,Longevity ,Homeostasis ,Insulin Resistance ,Mice ,Male ,Diet ,High-Fat ,Mice ,Inbred C57BL ,Mitochondria ,Cell Proliferation ,Mitophagy ,Insulin ,Gene Regulatory Networks - Abstract
Caloric restriction (CR) can extend the organism life- and health-span by improving glucose homeostasis. How CR affects the structure-function of pancreatic beta cells remains unknown. We used single nucleus transcriptomics to show that CR increases the expression of genes for beta cell identity, protein processing, and organelle homeostasis. Gene regulatory network analysis reveal that CR activates transcription factors important for beta cell identity and homeostasis, while imaging metabolomics demonstrates that beta cells upon CR are more energetically competent. In fact, high-resolution microscopy show that CR reduces beta cell mitophagy to increase mitochondria mass and the potential for ATP generation. However, CR beta cells have impaired adaptive proliferation in response to high fat diet feeding. Finally, we show that long-term CR delays the onset of beta cell aging hallmarks and promotes cell longevity by reducing beta cell turnover. Therefore, CR could be a feasible approach to preserve compromised beta cell structure-function during aging and diabetes.
- Published
- 2024
4. Dietary Anthocyanins Mitigate High-Fat Diet-Induced Hippocampal Inflammation in Mice
- Author
-
Muhammad, Imani, Cremonini, Eleonora, Mathieu, Patricia, Adamo, Ana M, and Oteiza, Patricia I
- Subjects
Biomedical and Clinical Sciences ,Nutrition and Dietetics ,Genetics ,Neurosciences ,Prevention ,Dietary Supplements ,Obesity ,Nutrition ,Complementary and Integrative Health ,2.1 Biological and endogenous factors ,Oral and gastrointestinal ,Metabolic and endocrine ,Cardiovascular ,Animals ,Anthocyanins ,Diet ,High-Fat ,Male ,Hippocampus ,Mice ,Inbred C57BL ,Mice ,Inflammation ,Brain-Derived Neurotrophic Factor ,high-fat diet ,anthocyanidins ,BDNF ,neuroin ,endotoxemia ,hippocampus ,HPA axis ,C57BL/6J mice ,neuroinflammation ,Animal Production ,Food Sciences ,Nutrition & Dietetics ,Animal production ,Food sciences ,Nutrition and dietetics - Abstract
BackgroundObesity and consumption of high-fat diets (HFD) are associated with intestinal permeabilization and increased paracellular transport of endotoxins, which can promote neuroinflammation. Inflammation can affect the hypothalamic pituitary adrenal (HPA) axis, which controls responses to stress and downregulates the brain-derived neurotrophic factor (BDNF), which can promote anxiety and depression, conditions frequently found in obesity. We previously showed that consumption of anthocyanins (AC) mitigate HFD-induced insulin resistance, intestinal permeability, and inflammation.ObjectivesThis study investigated if a dietary supplementation with a cyanidin- and delphinidin-rich extract (CDRE) could counteract HFD/obesity-induced hippocampal inflammation in mice.MethodsC57BL/6J male mice were fed for 14 wk on one of the following diets: 1) a control diet containing 10% total calories from fat (C), 2) a control diet supplemented with 40 mg AC/kg body weight (BW) (CAC), 3) a HFD containing 60% total calories from fat (lard) (HF), or 4) the HFD supplemented with 2, 20, or 40 mg AC/kg BW (HFA2, HFA20, and HFA40, respectively). In plasma and in the hippocampus, parameters of neuroinflammation and the underlying cause (endotoxemia) and consequences (alterations to the HPA and BDNF downregulation) were measured.ResultsConsumption of the HFD caused endotoxemia. Accordingly, hippocampal Tlr4 mRNA levels were 110% higher in the HF group, which were both prevented by CDRE supplementation. Consumption of the HFD also caused: 1) microgliosis and increased expression of genes involved in neuroinflammation, that is, Iba-1, Nox4, Tnfα, and Il-1β, 2) alterations of HPA axis regulation, that is, with low expression of mineralocorticoid (MR) and glucocorticoid (GR) receptors; and 3) decreased Bdnf expression. Supplementation of HFD-fed mice with CDRE mitigated neuroinflammation, microgliosis, and MR and BDNF decreases.ConclusionsCDRE supplementation mitigates the negative effects associated with HFD consumption and obesity in mouse hippocampus, in part by decreasing inflammation, improving glucocorticoid metabolism, and upregulating BDNF.
- Published
- 2024
5. High-Fat Feeding Alters Circulating Triglyceride Composition: Roles of FFA Desaturation and ω-3 Fatty Acid Availability.
- Author
-
Shen, Tong, Oh, Youngtaek, Jeong, Shinwu, Cho, Suengmok, Fiehn, Oliver, and Youn, Jang
- Subjects
cardiovascular disease ,diabetes ,fat oxidation ,lipidomics analysis ,palmitoleic acid ,polyunsaturated fatty acids ,Animals ,Triglycerides ,Fatty Acids ,Omega-3 ,Diet ,High-Fat ,Rats ,Male ,Rats ,Wistar ,Fatty Acids ,Nonesterified ,Stearoyl-CoA Desaturase ,Hypertriglyceridemia ,Lipidomics - Abstract
Hypertriglyceridemia is a risk factor for type 2 diabetes and cardiovascular disease (CVD). Plasma triglycerides (TGs) are a key factor for assessing the risk of diabetes or CVD. However, previous lipidomics studies have demonstrated that not all TG molecules behave the same way. Individual TGs with different fatty acid compositions are regulated differentially under various conditions. In addition, distinct groups of TGs were identified to be associated with increased diabetes risk (TGs with lower carbon number [C#] and double-bond number [DB#]), or with decreased risk (TGs with higher C# and DB#). In this study, we examined the effects of high-fat feeding in rats on plasma lipid profiles with special attention to TG profiles. Wistar rats were maintained on either a low-fat (control) or high-fat diet (HFD) for 2 weeks. Plasma samples were obtained before and 2.5 h after a meal (n = 10 each) and subjected to lipidomics analyses. High-fat feeding significantly impacted circulating lipid profiles, with the most significant effects observed on TG profile. The effects of an HFD on individual TG species depended on DB# in their fatty acid chains; an HFD increased TGs with low DB#, associated with increased diabetes risk, but decreased TGs with high DB#, associated with decreased risk. These changes in TGs with an HFD were associated with decreased indices of hepatic stearoyl-CoA desaturase (SCD) activity, assessed from hepatic fatty acid profiles. Decreased SCD activity would reduce the conversion of saturated to monounsaturated fatty acids, contributing to the increases in saturated TGs or TGs with low DB#. In addition, an HFD selectively depleted ω-3 polyunsaturated fatty acids (PUFAs), contributing to the decreases in TGs with high DB#. Thus, an HFD had profound impacts on circulating TG profiles. Some of these changes were at least partly explained by decreased hepatic SCD activity and depleted ω-3 PUFA.
- Published
- 2024
6. Obesity Alters POMC and Kisspeptin Neuron Cross Talk Leading to Reduced Luteinizing Hormone in Male Mice.
- Author
-
Villa, Pedro, Ruggiero-Ruff, Rebecca, Jamieson, Bradley, Campbell, Rebecca, and Coss, Djurdjica
- Subjects
GnRH ,POMC ,high-fat diet ,kisspeptin ,obesity ,Animals ,Male ,Kisspeptins ,Obesity ,Luteinizing Hormone ,Mice ,Neurons ,Pro-Opiomelanocortin ,Gonadotropin-Releasing Hormone ,Mice ,Inbred C57BL ,Diet ,High-Fat - Abstract
Obesity is associated with hypogonadism in males, characterized by low testosterone and sperm number. Previous studies determined that these stem from dysregulation of hypothalamic circuitry that regulates reproduction, by unknown mechanisms. Herein, we used mice fed chronic high-fat diet, which mimics human obesity, to determine mechanisms of impairment at the level of the hypothalamus, in particular gonadotropin-releasing hormone (GnRH) neurons that regulate luteinizing hormone (LH), which then regulates testosterone. Consistent with obese humans, we demonstrated lower LH, and lower pulse frequency of LH secretion, but unchanged pituitary responsiveness to GnRH. LH pulse frequency is regulated by pulsatile GnRH secretion, which is controlled by kisspeptin. Peripheral and central kisspeptin injections, and DREADD-mediated activation of kisspeptin neurons, demonstrated that kisspeptin neurons were suppressed in obese mice. Thus, we investigated regulators of kisspeptin secretion. We determined that the LH response to NMDA was lower in obese mice, corresponding to fewer glutamate receptors in kisspeptin neurons, which may be critical for kisspeptin synchronization. Given that kisspeptin neurons also interact with anorexigenic POMC neurons, which are affected by obesity, we examined their cross talk, and determined that the LH response to either DREADD-mediated activation of POMC neurons or central injection of αMSH, a product of POMC, is abolished in obese mice. This was accompanied by diminished levels of αMSH receptor, MC4R, in kisspeptin neurons. Together, our studies determined that obesity leads to the downregulation of receptors that regulate kisspeptin neurons, which is associated with lower LH pulse frequency, leading to lower LH and hypogonadism.
- Published
- 2024
7. Beneficial metabolic effects of PAHSAs depend on the gut microbiota in diet-induced obese mice but not in chow-fed mice.
- Author
-
Lee, Jennifer, Wellenstein, Kerry, Rahnavard, Ali, Nelson, Andrew, Holter, Marlena, Cummings, Bethany, Yeliseyev, Vladimir, Castoldi, Angela, Clish, Clary, Bry, Lynn, Siegel, Dionicio, and Kahn, Barbara
- Subjects
Bacteroides thetaiotaomicron ,PAHSAs ,diet-induced obesity ,glucose metabolism ,gut microbiota ,Animals ,Male ,Female ,Mice ,Gastrointestinal Microbiome ,Obesity ,Diet ,High-Fat ,Insulin Resistance ,Mice ,Inbred C57BL ,Stearic Acids ,Palmitic Acid ,Feces ,Mice ,Obese - Abstract
Dietary lipids play an essential role in regulating the function of the gut microbiota and gastrointestinal tract, and these luminal interactions contribute to mediating host metabolism. Palmitic Acid Hydroxy Stearic Acids (PAHSAs) are a family of lipids with antidiabetic and anti-inflammatory properties, but whether the gut microbiota contributes to their beneficial effects on host metabolism is unknown. Here, we report that treating chow-fed female and male germ-free (GF) mice with PAHSAs improves glucose tolerance, but these effects are lost upon high fat diet (HFD) feeding. However, transfer of feces from PAHSA-treated, but not vehicle-treated, chow-fed conventional mice increases insulin sensitivity in HFD-fed GF mice. Thus, the gut microbiota is necessary for, and can transmit, the insulin-sensitizing effects of PAHSAs in HFD-fed GF male mice. Analyses of the cecal metagenome and lipidome of PAHSA-treated mice identified multiple lipid species that associate with the gut commensal Bacteroides thetaiotaomicron (Bt) and with insulin sensitivity resulting from PAHSA treatment. Supplementing live, and to some degree, heat-killed Bt to HFD-fed female mice prevented weight gain, reduced adiposity, improved glucose tolerance, fortified the colonic mucus barrier and reduced systemic inflammation compared to HFD-fed controls. These effects were not observed in HFD-fed male mice. Furthermore, ovariectomy partially reversed the beneficial Bt effects on host metabolism, indicating a role for sex hormones in mediating the Bt probiotic effects. Altogether, these studies highlight the fact that PAHSAs can modulate the gut microbiota and that the microbiota is necessary for the beneficial metabolic effects of PAHSAs in HFD-fed mice.
- Published
- 2024
8. Effects of beef fat enriched with trans vaccenic acid and cis9, trans11-CLA on glucose homoeostasis and hepatic lipid accumulation in high-fat diet-induced obese mice.
- Author
-
Xu, Yanqing, Hsu, Ming-Fo, Haj, Fawaz, and Vahmani, Payam
- Subjects
Beef ,Biohydrogenation ,Type 2 diabetes ,trans-FA ,Animals ,Male ,Diet ,High-Fat ,Mice ,Inbred C57BL ,Liver ,Obesity ,Homeostasis ,Oleic Acids ,Mice ,Cattle ,Red Meat ,Lipid Metabolism ,Linoleic Acids ,Conjugated ,Dietary Fats ,Glucose ,Mice ,Obese ,Adiposity ,Fatty Liver ,Blood Glucose ,Triglycerides - Abstract
Trans vaccenic acid (TVA, trans11-18 : 1) and cis9, trans11-CLA (also known as rumenic acid; RA) have received widespread attention as potentially beneficial trans-FA due to their putative health benefits, including anti-diabetic properties. The objective of this study was to determine the effects of beef fat naturally enriched with TVA and RA on parameters related to glucose homoeostasis and associated metabolic markers in diet-induced obese (DIO) mice. Thirty-six male C57BL/6J mice (8 weeks old) were fed for 19 weeks with either a control low-fat diet (CLF), a control high-fat diet (CHF), or a TVA+RA-enriched high-fat diet (EHF). Compared with CLF, feeding either CHF or EHF resulted in adverse metabolic outcomes associated with high-fat diets, including adiposity, impaired glucose control and hepatic steatosis. However, the EHF diet induced a significantly higher liver weight TAG content and elevated plasma alanine transaminase levels compared with the CHF diet. Collectively, the findings from this study suggest that EHF does not improve glucose tolerance and worsens liver steatosis in DIO mice. However, the adverse effects of EHF on the liver could be in part related to the presence of other trans-FA in the enriched beef fat.
- Published
- 2024
9. Cholinergic Neurotransmission Controls Orexigenic Endocannabinoid Signaling in the Gut in Diet-Induced Obesity.
- Author
-
Wood, Courtney P, Alvarez, Camila, and DiPatrizio, Nicholas V
- Subjects
Pharmacology and Pharmaceutical Sciences ,Biomedical and Clinical Sciences ,Digestive Diseases ,Neurosciences ,Obesity ,Nutrition ,Endocannabinoid System Research ,Cannabinoid Research ,2.1 Biological and endogenous factors ,Oral and gastrointestinal ,Animals ,Endocannabinoids ,Male ,Mice ,Synaptic Transmission ,Mice ,Inbred C57BL ,Diet ,High-Fat ,Signal Transduction ,Glycerides ,Arachidonic Acids ,Eating ,Muscarinic Antagonists ,Receptors ,Muscarinic ,Brain-Gut Axis ,cholinergic ,endocannabinoid ,gut–brain ,intestine ,obesity ,parasympathetic ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Neurology & Neurosurgery - Abstract
The brain bidirectionally communicates with the gut to control food intake and energy balance, which becomes dysregulated in obesity. For example, endocannabinoid (eCB) signaling in the small-intestinal (SI) epithelium is upregulated in diet-induced obese (DIO) mice and promotes overeating by a mechanism that includes inhibiting gut-brain satiation signaling. Upstream neural and molecular mechanism(s) involved in overproduction of orexigenic gut eCBs in DIO, however, are unknown. We tested the hypothesis that overactive parasympathetic signaling at the muscarinic acetylcholine receptors (mAChRs) in the SI increases biosynthesis of the eCB, 2-arachidonoyl-sn-glycerol (2-AG), which drives hyperphagia via local CB1Rs in DIO. Male mice were maintained on a high-fat/high-sucrose Western-style diet for 60 d, then administered several mAChR antagonists 30 min prior to tissue harvest or a food intake test. Levels of 2-AG and the activity of its metabolic enzymes in the SI were quantitated. DIO mice, when compared to those fed a low-fat/no-sucrose diet, displayed increased expression of cFos protein in the dorsal motor nucleus of the vagus, which suggests an increased activity of efferent cholinergic neurotransmission. These mice exhibited elevated levels of 2-AG biosynthesis in the SI, that was reduced to control levels by mAChR antagonists. Moreover, the peripherally restricted mAChR antagonist, methylhomatropine bromide, and the peripherally restricted CB1R antagonist, AM6545, reduced food intake in DIO mice for up to 24 h but had no effect in mice conditionally deficient in SI CB1Rs. These results suggest that hyperactivity at mAChRs in the periphery increases formation of 2-AG in the SI and activates local CB1Rs, which drives hyperphagia in DIO.
- Published
- 2024
10. Protaetia brevitarsis Hydrolysate Mitigates Muscle Dysfunction and Ectopic Fat Deposition Triggered by a High-Fat Diet in Mice.
- Author
-
Park, Kyungeun, Jung, Sunyoon, Ha, Jung-Heun, and Jeong, Yoonhwa
- Abstract
Background/Objectives: Obesity is a key factor in metabolic syndrome (MetS) development. Consumption of a high-fat diet (HFD) accelerates the onset of obesity and associated metabolic complications. Protaetia brevitarsis (PB) has been traditionally utilized in Korean medicine for its antioxidant, anti-diabetic, anticancer, and hepatoprotective effects. However, specific effects of PB hydrolysate on skeletal muscles have not been fully elucidated. Therefore, this study sought to assess the influence of PB on HFD-induced MetS, focusing on the lipid metabolism and inflammatory responses mediated by AMP-activated protein kinase activation. Methods: To induce obesity, 6-week-old C57BL/6J mice were maintained on an HFD for 8 weeks, after which PB hydrolysate was orally administered for 16 weeks while the HFD regimen was sustained. A glucose tolerance test was conducted orally to evaluate glucose regulation, and forelimb grip strength was assessed upon completion of the experimental period. Histological assessments, serum biochemical analysis, lipid extraction, Western blot analysis, and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) were performed following euthanasia. Results: PB significantly reduced ectopic lipid deposition in skeletal muscles, enhanced muscle strength, and improved insulin sensitivity by increasing fatty acid oxidation via AMP-activated protein kinase/carnitine palmitoyltransferase 1 activation and inhibiting lipogenesis via stearoyl-CoA desaturase 1 gene downregulation. Furthermore, PB alleviated HFD-induced low-grade chronic inflammation by decreasing systemic monocyte chemoattractant protein 1 levels, thereby reducing ectopic fat deposition. Conclusions: This study highlights the potential of PB as a nutraceutical to mitigate MetS in HFD-fed mice. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
11. Nice to know 2: The impact of NICE guidelines on ketogenic diet services in the UK and Ireland – An update.
- Author
-
Whiteley, Victoria J. and Schoeler, Natasha E.
- Subjects
- *
DIETITIANS' attitudes , *MEDICAL protocols , *KETOGENIC diet , *THERAPEUTICS , *DESCRIPTIVE statistics , *TREATMENT duration , *LOW-carbohydrate diet , *SURVEYS , *EPILEPSY , *RESEARCH , *CLINICAL deterioration , *NUTRITION services , *COMPARATIVE studies , *TRIGLYCERIDES - Abstract
Background: Ketogenic diet therapy (KDT) has been recommended as a treatment for drug‐resistant epilepsy in children and young people since 2012 in the National Institute for Health and Care Excellence Clinical Guidelines for Epilepsies. The Ketogenic Dietitians Research Network completed a survey in 2017 to assess the impact of these guidelines. Methods: An online survey was circulated to ketogenic dietitians across the UK and Ireland. The results were compared with those of the 2017 survey. Results: The number of individuals following KDT was 854, comprising an increase of 13% since 2017. Service sizes ranged widely, with 1–74 (median 16) patients on the diet. Of 36 services, 30 had a waiting list, ranging from 2 to 67 (median 9) patients. The classical diet continued to be the most common KDT used (58% of patients). Ten services reported use of a new flexible medium chain triglyceride protocol. Some 48% of patients (n = 427) had been following the KDT for over 2 years, comprising an 18% increase since 2017. Of these, 68 (15.9%) had attempted to wean off KDT but had to re‐start as a result of a deterioration in seizures. Conclusions: The number of individuals following medical KDT remains stable. Referral numbers and waiting lists remain high, highlighting that KDT is still a well‐recognised treatment option for drug‐resistant epilepsy. The types of KDT used are similar to previous years, although increasingly flexible protocols are being adopted. Longer‐term use of KDT is increasing, with a proportion of patients requiring long‐term use to maintain seizure control. Highlights: Ketogenic diets remain a well‐recognised treatment option for drug‐resistant epilepsy in the UK and Ireland with the introduction of more flexible protocols in adults and children. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Sexual Dimorphism in the Immunometabolic Role of Gpr183 in Mice.
- Author
-
Voss, Liv von, Arora, Tulika, Assis, Juliana, Kuentzel, Katharina B, Arfelt, Kristine N, Nøhr, Mark K, Grevengoed, Trisha J, Arumugam, Manimozhiyan, Mandrup-Poulsen, Thomas, and Rosenkilde, Mette M
- Subjects
SEXUAL dimorphism ,INFLAMMATORY bowel diseases ,G protein coupled receptors ,BODY composition ,WESTERN diet - Abstract
Context Excessive eating and intake of a Western diet negatively affect the intestinal immune system, resulting in compromised glucose homeostasis and lower gut bacterial diversity. The G protein–coupled receptor GPR183 regulates immune cell migration and intestinal immune response and has been associated with tuberculosis, type 1 diabetes, and inflammatory bowel diseases. Objective We hypothesized that with these implications, GPR183 has an important immunometabolic role and investigated this using a global Gpr183 knockout mouse model. Methods Wild-type (WT) and Gpr183 -deficient (Gpr183
–/– ) mice were fed a high-fat, high-sucrose diet (HFSD) for 15 weeks. We investigated changes in weight, body composition, fecal immunoglobulin A (IgA) levels, fecal microbiome, and glucose tolerance before and after the diet. Macrophage infiltration into visceral fat was determined by flow cytometry, and hepatic gene expression was measured. Results A sexual dimorphism was discovered, whereby female Gpr183–/– mice showed adverse metabolic outcomes compared to WT counterparts with inferior glucose tolerance, lower fecal IgA levels, and increased macrophage infiltration in visceral fat. In contrast, male Gpr183–/– mice had significantly lower fasting blood glucose after diet than male WT mice. Liver gene expression showed reduced inflammation and macrophage markers in Gpr183–/– livers, regardless of sex, while the pancreatic islet area did not differ between the groups. No conclusive differences were found after microbiome sequencing. Conclusion Gpr183 maintains metabolic homeostasis in female but not in male mice independent of diet. If confirmed in humans, future therapy targeting GPR183 should consider this sexual dimorphism. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
13. The Role of Gut Microbiota in Male Erectile Dysfunction of Rats
- Author
-
Zhunan Xu, Shangren Wang, Chunxiang Liu, Jiaqi Kang, Yang Pan, Zhexin Zhang, Hang Zhou, Mingming Xu, Xia Li, Haoyu Wang, Shuai Niu, Li Liu, Daqing Sun, and Xiaoqiang Liu
- Subjects
diet ,high-fat ,erectile dysfunction ,gastrointestinal microbiome ,inflammation ,metabolomics ,Medicine ,Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
Purpose: Erectile dysfunction (ED) is a common male sexual dysfunction. Gut microbiota plays an important role in various diseases. To investigate the effects and mechanisms of intestinal flora dysregulation induced by high-fat diet (HFD) on erectile function. Materials and Methods: Male Sprague–Dawley rats aged 8 weeks were randomly divided into the normal diet (ND) and HFD groups. After 24 weeks, a measurement of erectile function was performed. We performed 16S rRNA sequencing of stool samples. Then, we established fecal microbiota transplantation (FMT) rat models by transplanting fecal microbiota from rats of ND group and HFD group to two new groups of rats respectively. After 24 weeks, erectile function of the rats was evaluated and 16S rRNA sequencing was performed, and serum samples were collected for the untargeted metabolomics detection. Results: The erectile function of rats and the species diversity of intestinal microbiota in the HFD group was significantly lower, and the characteristics of the intestinal microbiota community structure were also significantly different between the two groups. The erectile function of rats in the HFD-FMT group was significantly lower than that of rats in the ND-FMT group. The characteristics of the intestinal microbiota community structure were significantly different. In the HFD-FMT group, 27 metabolites were significantly different and they were mainly involved in the several inflammation-related pathways. Conclusions: Intestinal microbiota disorders induced by HFD can damage the intestinal barrier of rats, change the serum metabolic profile, induce low-grade inflammation and apoptosis in the corpus cavernosum of the penis, and lead to ED.
- Published
- 2025
- Full Text
- View/download PDF
14. Exercise mitigates flow recirculation and activates metabolic transducer SCD1 to catalyze vascular protective metabolites.
- Author
-
Cavallero, Susana, Roustaei, Mehrdad, Satta, Sandro, Cho, Jae, Phan, Henry, Baek, Kyung, Blázquez-Medela, Ana, Gonzalez-Ramos, Sheila, Vu, Khoa, Park, Seul-Ki, Yokota, Tomohiro, Mack, Julia, Sigmund, Curt, Reddy, Srinivasa, Li, Rongsong, Hsiai, Tzung, and Sumner, Jennifer
- Subjects
Animals ,Mice ,Aorta ,Diet ,High-Fat ,Endothelium ,Vascular ,Motor Activity ,Stearoyl-CoA Desaturase - Abstract
Exercise promotes pulsatile shear stress in the arterial circulation and ameliorates cardiometabolic diseases. However, exercise-mediated metabolic transducers for vascular protection remain under-investigated. Untargeted metabolomic analysis demonstrated that wild-type mice undergoing voluntary wheel running exercise expressed increased endothelial stearoyl-CoA desaturase 1 (SCD1) that catalyzes anti-inflammatory lipid metabolites, namely, oleic (OA) and palmitoleic acids (PA), to mitigate NF-κB-mediated inflammatory responses. In silico analysis revealed that exercise augmented time-averaged wall shear stress but mitigated flow recirculation and oscillatory shear index in the lesser curvature of the mouse aortic arch. Following exercise, endothelial Scd1-deleted mice (Ldlr-/- Scd1EC-/-) on high-fat diet developed persistent VCAM1-positive endothelium in the lesser curvature and the descending aorta, whereas SCD1 overexpression via adenovirus transfection mitigated endoplasmic reticulum stress and inflammatory biomarkers. Single-cell transcriptomics of the aorta identified Scd1-positive and Vcam1-negative endothelial subclusters interacting with other candidate genes. Thus, exercise mitigates flow recirculation and activates endothelial SCD1 to catalyze OA and PA for vascular endothelial protection.
- Published
- 2024
15. The Impact of a Ketogenic Diet on Late-Stage Pancreatic Carcinogenesis in Mice: Efficacy and Safety Studies
- Author
-
Cortez, Natalia E, Bacha, Tarek A, Ead, Aya Samir, Lanzi, Cecilia Rodriguez, Lacroix, Cassandra, Franceschetti, Anais, Hong, Brian V, Matsukuma, Karen, and Mackenzie, Gerardo G
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Prevention ,Digestive Diseases ,Complementary and Integrative Health ,Rare Diseases ,Pancreatic Cancer ,Obesity ,Nutrition ,Liver Disease ,Oral and gastrointestinal ,Animals ,Diet ,Ketogenic ,Pancreatic Neoplasms ,Male ,Female ,Diet ,High-Fat ,Mice ,Carcinogenesis ,Liver ,Leptin ,Insulin ,Disease Models ,Animal ,Pancreas ,pancreatic cancer ,ketogenic diet ,high-fat diet ,pancreatic carcinogenesis ,Food Sciences ,Nutrition and Dietetics ,Clinical sciences ,Nutrition and dietetics ,Public health - Abstract
BackgroundHigh-fat diets (HFDs) have been associated with an increased risk of pancreatic cancer. In contrast, ketogenic diets (KDs) have been shown to display anti-tumor characteristics. The objective of this work was to evaluate the efficacy of a KD on late-stage pancreatic carcinogenesis in a genetically modified mouse model of pancreatic cancer [LSL-KrasG12D/+; Ptf1-Cre (KC) mice], as well as its liver safety, and to compare it to that of an HFD.MethodsSix-month-old female and male KC mice were randomly allocated to either a control diet (CD) (%kcal: 20% fat, 15% protein, 65% carbohydrates), an HFD (%kcal: 40% fat, 15% protein, 45% carbohydrate) or a KD (%kcal: 84% fat, 15% protein, 1% carbohydrate) and fed these diets for 6 months.ResultsHFD-fed, but not KD-fed, mice showed a 15% increase in body weight, plus elevated serum insulin (2.4-fold increase) and leptin (2.9-fold increase) levels, compared to CD-fed mice. At the pancreas level, no differences in pancreatic cancer incidence rates were observed among the diet groups. Regarding the liver safety profile, the HFD-fed mice had higher serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), and alkaline phosphatase (ALP), when compared to the CD and KD groups. In addition, upon histologic examination, an HFD, but not a KD, showed a ~2-fold increase in both macro- and microsteatosis, as well as 35% and 32% higher levels of TLR4 and NF-κB activation, respectively, compared to CD-fed mice.ConclusionsIn summary, although a KD intervention alone did not prevent pancreatic carcinogenesis, our data suggests that a KD modulates insulin signaling and hepatic lipid metabolism, highlighting its beneficial effects on healthspan and liver function when compared to an HFD.
- Published
- 2024
16. Anti-Inflammatory Oxysterol, Oxy210, Inhibits Atherosclerosis in Hyperlipidemic Mice and Inflammatory Responses of Vascular Cells
- Author
-
Stappenbeck, Frank, Wang, Feng, Sinha, Satyesh K, Hui, Simon T, Farahi, Lia, Mukhamedova, Nigora, Fleetwood, Andrew, Murphy, Andrew J, Sviridov, Dmitri, Lusis, Aldons J, and Parhami, Farhad
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Liver Disease ,Atherosclerosis ,Hepatitis ,Cardiovascular ,Aging ,Digestive Diseases ,Nutrition ,2.1 Biological and endogenous factors ,Animals ,Mice ,Humans ,Inflammation ,Endothelial Cells ,Oxysterols ,Anti-Inflammatory Agents ,Hyperlipidemias ,Macrophages ,Male ,Disease Models ,Animal ,Diet ,High-Fat ,atherosclerosis ,NAFLD/NASH ,MAFLD/MASH ,oxysterols ,Oxy210 ,APOE*3-Leiden.CETP mouse model ,chronic inflammation ,fibrosis ,endothelial cells ,macrophages ,Biological sciences ,Biomedical and clinical sciences - Abstract
Background and aimsWe previously reported that Oxy210, an oxysterol-based drug candidate, exhibits antifibrotic and anti-inflammatory properties. We also showed that, in mice, it ameliorates hepatic hallmarks of non-alcoholic steatohepatitis (NASH), including inflammation and fibrosis, and reduces adipose tissue inflammation. Here, we aim to investigate the effects of Oxy210 on atherosclerosis, an inflammatory disease of the large arteries that is linked to NASH in epidemiologic studies, shares many of the same risk factors, and is the major cause of mortality in people with NASH.MethodsOxy210 was studied in vivo in APOE*3-Leiden.CETP mice, a humanized mouse model for both NASH and atherosclerosis, in which symptoms are induced by consumption of a high fat, high cholesterol "Western" diet (WD). Oxy210 was also studied in vitro using two cell types that are important in atherogenesis: human aortic endothelial cells (HAECs) and macrophages treated with atherogenic and inflammatory agents.ResultsOxy210 reduced atherosclerotic lesion formation by more than 50% in hyperlipidemic mice fed the WD for 16 weeks. This was accompanied by reduced plasma cholesterol levels and reduced macrophages in lesions. In HAECs and macrophages, Oxy210 reduced the expression of key inflammatory markers associated with atherosclerosis, including interleukin-1 beta (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), chemokine (C-C motif) ligand 2 (CCL2), vascular cell adhesion molecule-1 (VCAM-1), and E-Selectin. In addition, cholesterol efflux was significantly enhanced in macrophages treated with Oxy210.ConclusionsThese findings suggest that Oxy210 could be a drug candidate for targeting both NASH and atherosclerosis, as well as chronic inflammation associated with the manifestations of metabolic syndrome.
- Published
- 2024
17. Sympathetic innervation of interscapular brown adipose tissue is not a predominant mediator of oxytocin-elicited reductions of body weight and adiposity in male diet-induced obese mice
- Author
-
Edwards, Melise M, Nguyen, Ha K, Dodson, Andrew D, Herbertson, Adam J, Wolden-Hanson, Tami, Wietecha, Tomasz A, Honeycutt, Mackenzie K, Slattery, Jared D, O’Brien, Kevin D, Graham, James L, Havel, Peter J, Mundinger, Thomas O, Sikkema, Carl L, Peskind, Elaine R, Ryu, Vitaly, Taborsky, Gerald J, and Blevins, James E
- Subjects
Medical Physiology ,Biomedical and Clinical Sciences ,Nutrition ,Obesity ,Neurosciences ,Metabolic and endocrine ,Animals ,Oxytocin ,Adipose Tissue ,Brown ,Male ,Mice ,Sympathetic Nervous System ,Diet ,High-Fat ,Adiposity ,Mice ,Inbred C57BL ,Body Weight ,Weight Loss ,Mice ,Obese ,Energy Metabolism ,Norepinephrine ,obesity ,brown adipose tissue ,white adipose tissue ,oxytocin ,food intake ,Clinical Sciences ,Nutrition and Dietetics ,Clinical sciences - Abstract
Previous studies indicate that CNS administration of oxytocin (OT) reduces body weight in high fat diet-induced obese (DIO) rodents by reducing food intake and increasing energy expenditure (EE). We recently demonstrated that hindbrain (fourth ventricular [4V]) administration of OT elicits weight loss and elevates interscapular brown adipose tissue temperature (TIBAT, a surrogate measure of increased EE) in DIO mice. What remains unclear is whether OT-elicited weight loss requires increased sympathetic nervous system (SNS) outflow to IBAT. We hypothesized that OT-induced stimulation of SNS outflow to IBAT contributes to its ability to activate BAT and elicit weight loss in DIO mice. To test this hypothesis, we determined the effect of disrupting SNS activation of IBAT on the ability of 4V OT administration to increase TIBAT and elicit weight loss in DIO mice. We first determined whether bilateral surgical SNS denervation to IBAT was successful as noted by ≥ 60% reduction in IBAT norepinephrine (NE) content in DIO mice. NE content was selectively reduced in IBAT at 1-, 6- and 7-weeks post-denervation by 95.9 ± 2.0, 77.4 ± 12.7 and 93.6 ± 4.6% (P
- Published
- 2024
18. Neurodevelopment Is Dependent on Maternal Diet: Placenta and Brain Glucose Transporters GLUT1 and GLUT3
- Author
-
Daida, Tomoko, Shin, Bo-Chul, Cepeda, Carlos, and Devaskar, Sherin U
- Subjects
Medical Physiology ,Biomedical and Clinical Sciences ,Intellectual and Developmental Disabilities (IDD) ,Neurosciences ,Mental Health ,Pediatric ,Behavioral and Social Science ,Nutrition ,Brain Disorders ,Autism ,Diabetes ,Complementary and Integrative Health ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Neurological ,Reproductive health and childbirth ,Glucose Transporter Type 3 ,Humans ,Pregnancy ,Brain ,Placenta ,Female ,Glucose Transporter Type 1 ,Neurodevelopmental Disorders ,Animals ,Maternal Nutritional Physiological Phenomena ,Diet ,Ketogenic ,Diet ,High-Fat ,Neurodegenerative Diseases ,Glucose ,glucose transporter ,glucose transporter 3 ,maternal diet ,postnatal diet ,neurodevelopment ,neurodevelopmental disorders ,neurodegenerative disorders ,Huntington's disease ,brain ,placenta ,Huntington’s disease ,Food Sciences ,Nutrition and Dietetics ,Clinical sciences ,Nutrition and dietetics ,Public health - Abstract
Glucose is the primary energy source for most mammalian cells and its transport is affected by a family of facilitative glucose transporters (GLUTs) encoded by the SLC2 gene. GLUT1 and GLUT3, highly expressed isoforms in the blood-brain barrier and neuronal membranes, respectively, are associated with multiple neurodevelopmental disorders including epilepsy, dyslexia, ADHD, and autism spectrum disorder (ASD). Dietary therapies, such as the ketogenic diet, are widely accepted treatments for patients with the GLUT1 deficiency syndrome, while ameliorating certain symptoms associated with GLUT3 deficiency in animal models. A ketogenic diet, high-fat diet, and calorie/energy restriction during prenatal and postnatal stages can also alter the placental and brain GLUTs expression with long-term consequences on neurobehavior. This review focuses primarily on the role of diet/energy perturbations upon GLUT isoform-mediated emergence of neurodevelopmental and neurodegenerative disorders.
- Published
- 2024
19. Glucocorticoid signaling and the impact of high-fat diet on adipogenesis in vivo
- Author
-
Babel, Noah K and Feldman, Brian J
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Obesity ,Nutrition ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Humans ,Adipogenesis ,Glucocorticoids ,Diet ,High-Fat ,Adipocytes ,Endocrinology & Metabolism ,Clinical sciences - Abstract
Our research used glucocorticoids as a medically relevant molecular probe to identify a previously unrecognized ADAMTS1-PTN-Wnt pathway. We elucidated the role of this pathway in regulating adipose precursor cell (APC) behavior to either proliferate or differentiate in response to systemic cues, such as elevated caloric intake. Further, our studies identified the non-muscle myosin protein MYH9 as a key target of this pathway to modulate adipogenesis in vivo. These findings enable strategies toward developing novel therapeutics for obesity and related metabolic disorders.
- Published
- 2024
20. A Maternal Western-Style Diet Impairs Skeletal Muscle Lipid Metabolism in Adolescent Japanese Macaques.
- Author
-
Greyslak, Keenan, Hetrick, Byron, Bergman, Bryan, Dean, Tyler, Wesolowski, Stephanie, Gannon, Maureen, Schenk, Simon, Sullivan, Elinor, Aagaard, Kjersti, Kievit, Paul, Chicco, Adam, Friedman, Jacob, and McCurdy, Carrie
- Subjects
Humans ,Animals ,Pregnancy ,Female ,Adolescent ,Insulin Resistance ,Macaca fuscata ,Lipid Metabolism ,Muscle ,Skeletal ,Insulin ,Diet ,Western ,Fatty Acids ,Ceramides ,Diet ,High-Fat - Abstract
UNLABELLED: Maternal consumption of a Western-style diet (mWD) during pregnancy alters fatty acid metabolism and reduces insulin sensitivity in fetal skeletal muscle. The long-term impact of these fetal adaptations and the pathways underlying disordered lipid metabolism are incompletely understood. Therefore, we tested whether a mWD chronically fed to lean, insulin-sensitive adult Japanese macaques throughout pregnancy and lactation would impact skeletal muscle oxidative capacity and lipid metabolism in adolescent offspring fed a postweaning (pw) Western-style diet (WD) or control diet (CD). Although body weight was not different, retroperitoneal fat mass and subscapular skinfold thickness were significantly higher in pwWD offspring consistent with elevated fasting insulin and glucose. Maximal complex I (CI)-dependent respiration in muscle was lower in mWD offspring in the presence of fatty acids, suggesting that mWD impacts muscle integration of lipid with nonlipid oxidation. Abundance of all five oxidative phosphorylation complexes and VDAC, but not ETF/ETFDH, were reduced with mWD, partially explaining the lower respiratory capacity with lipids. Muscle triglycerides increased with pwWD; however, the fold increase in lipid saturation, 1,2-diacylglycerides, and C18 ceramide compared between pwCD and pwWD was greatest in mWD offspring. Reductions in CI abundance and VDAC correlated with reduced markers of oxidative stress, suggesting that these reductions may be an early-life adaptation to mWD to mitigate excess reactive oxygen species. Altogether, mWD, independent of maternal obesity or insulin resistance, results in sustained metabolic reprogramming in offspring muscle despite a healthy diet intervention. ARTICLE HIGHLIGHTS: In lean, active adolescent offspring, a postweaning Western-style diet (pwWD) leads to shifts in body fat distribution that are associated with poorer insulin sensitivity. Fatty acid-linked oxidative metabolism was reduced in skeletal muscles from offspring exposed to maternal Western-style diet (mWD) even when weaned to a healthy control diet for years. Reduced oxidative phosphorylation complex I-V and VDAC1 abundance partially explain decreased skeletal muscle respiration in mWD offspring. Prior exposure to mWD results in greater fold increase with pwWD in saturated lipids and bioactive lipid molecules (i.e. ceramide and sphingomyelin) associated with insulin resistance.
- Published
- 2023
21. Effects of high-fat, low-carbohydrate enteral nutrition in critically ill patients: A systematic review with meta-analysis.
- Author
-
Ohbe, Hiroyuki, Yoshida, Minoru, Okada, Kazuya, Inoue, Takaaki, Yamada, Kohei, Nakamura, Kensuke, Yamamoto, Ryo, Nozaki, Ayumu, Higashibeppu, Naoki, and Kotani, Joji
- Abstract
High-fat, low-carbohydrate enteral nutrition has gained attention, with expectations of an improved respiratory condition, fewer complications, and lower mortality. The present study performed a systematic review and meta-analysis of randomized controlled trials to examine the effects of high-fat, low-carbohydrate enteral nutrition in critically ill adult patients. We searched MEDLINE via Pubmed, Cochrane Central Register of Controlled Trials (CENTRAL), and ICHUSHI for randomized controlled trials comparing high-fat, low-carbohydrate enteral nutrition to standard enteral nutrition in critically ill adult patients who received enteral nutrition. The primary outcome was mortality. Secondary outcomes included intensive care unit (ICU) mortality, length of ICU stay, length of mechanical ventilation, and adverse events of diarrhea and gastric residual volume. We examined the risk of bias using the Cochrane risk-of-bias tool for randomized trials version 2. We assessed the overall certainty of evidence based on the Grading of Recommendations Assessment, Development, and Evaluation methodology. Synthesis results were calculated with risk ratios and 95% confidence intervals using a Mantel-Haenszel random-effects model. Eight trials with 607 patients were included. The effects of high-fat, low-carbohydrate enteral nutrition on mortality did not significantly differ from those of standard enteral nutrition (62/280 [22.1%] vs. 39/207 [18.8%], risk ratios = 1.14, 95% confidence intervals 0.80 to 1.62, P = 0.47). No significant differences were observed in ICU mortality, ICU length of stay, diarrhea, or gastric residual volume between the two groups. However, high-fat, low-carbohydrate enteral nutrition was associated with a significantly shorter duration of mechanical ventilation (mean difference −1.72 days, 95% confidence intervals −2.93 to −0.50, P = 0.005). High-fat, low-carbohydrate enteral nutrition may not affect mortality, but may decrease the duration of mechanical ventilation in critically ill adult patients. Limitations include the small number of studies and potential for bias. Further research is needed to confirm these results and investigate effects on other outcomes and in a subgroup of patients requiring mechanical ventilation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Precision pharmacological reversal of strain-specific diet-induced metabolic syndrome in mice informed by epigenetic and transcriptional regulation.
- Author
-
Wulfridge, Phillip, Davidovich, Adam, Salvador, Anna, Manno, Gabrielle, Tryggvadottir, Rakel, Idrizi, Adrian, Huda, M, Adams, L, Hansen, Kasper, Threadgill, David, Feinberg, Andrew, and Bennett, Brian
- Subjects
Humans ,Mice ,Animals ,Metabolic Syndrome ,Epigenomics ,Pilot Projects ,Liver ,Mice ,Inbred C57BL ,Mice ,Inbred NOD ,Diet ,High-Fat ,Epigenesis ,Genetic - Abstract
Diet-related metabolic syndrome is the largest contributor to adverse health in the United States. However, the study of gene-environment interactions and their epigenomic and transcriptomic integration is complicated by the lack of environmental and genetic control in humans that is possible in mouse models. Here we exposed three mouse strains, C57BL/6J (BL6), A/J, and NOD/ShiLtJ (NOD), to a high-fat, high-carbohydrate diet, leading to varying degrees of metabolic syndrome. We then performed transcriptomic and genome-wide DNA methylation analyses for each strain and found overlapping but also highly divergent changes in gene expression and methylation upstream of the discordant metabolic phenotypes. Strain-specific pathway analysis of dietary effects revealed a dysregulation of cholesterol biosynthesis common to all three strains but distinct regulatory networks driving this dysregulation. This suggests a strategy for strain-specific targeted pharmacologic intervention of these upstream regulators informed by epigenetic and transcriptional regulation. As a pilot study, we administered the drug GW4064 to target one of these genotype-dependent networks, the farnesoid X receptor pathway, and found that GW4064 exerts strain-specific protection against dietary effects in BL6, as predicted by our transcriptomic analysis. Furthermore, GW4064 treatment induced inflammatory-related gene expression changes in NOD, indicating a strain-specific effect in its associated toxicities as well as its therapeutic efficacy. This pilot study demonstrates the potential efficacy of precision therapeutics for genotype-informed dietary metabolic intervention and a mouse platform for guiding this approach.
- Published
- 2023
23. Obesity-associated microglial inflammatory activation paradoxically improves glucose tolerance.
- Author
-
Douglass, John, Ness, Kelly, Valdearcos, Martin, Wyse-Jackson, Alice, Dorfman, Mauricio, Frey, Jeremy, Fasnacht, Rachael, Santiago, Olivia, Niraula, Anzela, Banerjee, Jineta, Robblee, Megan, Koliwad, Suneil, and Thaler, Joshua
- Subjects
POMC ,TNF ,chemogenetic ,glucose sensing ,glucose tolerance ,hypothalamus ,insulin ,microglia ,obesity ,parasympathetic ,Humans ,Microglia ,NF-kappa B ,Obesity ,Body Weight ,Glucose ,Hypothalamus ,Diet ,High-Fat - Abstract
Hypothalamic gliosis associated with high-fat diet (HFD) feeding increases susceptibility to hyperphagia and weight gain. However, the body-weight-independent contribution of microglia to glucose regulation has not been determined. Here, we show that reducing microglial nuclear factor κB (NF-κB) signaling via cell-specific IKKβ deletion exacerbates HFD-induced glucose intolerance despite reducing body weight and adiposity. Conversely, two genetic approaches to increase microglial pro-inflammatory signaling (deletion of an NF-κB pathway inhibitor and chemogenetic activation through a modified Gq-coupled muscarinic receptor) improved glucose tolerance independently of diet in both lean and obese rodents. Microglial regulation of glucose homeostasis involves a tumor necrosis factor alpha (TNF-α)-dependent mechanism that increases activation of pro-opiomelanocortin (POMC) and other hypothalamic glucose-sensing neurons, ultimately leading to a marked amplification of first-phase insulin secretion via a parasympathetic pathway. Overall, these data indicate that microglia regulate glucose homeostasis in a body-weight-independent manner, an unexpected mechanism that limits the deterioration of glucose tolerance associated with obesity.
- Published
- 2023
24. Adipocyte-Derived PXR Signaling Is Dispensable for Diet-Induced Obesity and Metabolic Disorders in Mice.
- Author
-
Wang, Fang, Liu, Jingwei, Hernandez, Rebecca, Park, Se-Hyung, Lai, Ying-Jing, Wang, Shuxia, Blumberg, Bruce, and Zhou, Changcheng
- Subjects
Male ,Mice ,Animals ,Pregnane X Receptor ,Receptors ,Steroid ,Xenobiotics ,Obesity ,Adipocytes ,Insulin Resistance ,Diet ,High-Fat - Abstract
Pregnane X receptor (PXR) is a xenobiotic receptor that can be activated by numerous chemicals including endogenous hormones, dietary steroids, pharmaceutical agents, and environmental chemicals. PXR has been established to function as a xenobiotic sensor to coordinately regulate xenobiotic metabolism by regulating the expression of many enzymes and transporters required for xenobiotic metabolism. Recent studies have implicated a potentially important role for PXR in obesity and metabolic disease beyond xenobiotic metabolism, but how PXR action in different tissues or cell types contributes to obesity and metabolic disorders remains elusive. To investigate the role of adipocyte PXR in obesity, we generated a novel adipocyte-specific PXR deficient mouse model (PXRΔAd). Notably, we found that loss of adipocyte PXR did not affect food intake, energy expenditure, and obesity in high-fat diet-fed male mice. PXRΔAd mice also had similar obesity-associated metabolic disorders including insulin resistance and hepatic steatosis as control littermates. PXR deficiency in adipocytes did not affect expression of key adipose genes in PXRΔAd mice. Our findings suggest that adipocyte PXR signaling may be dispensable in diet-induced obesity and metabolic disorders in mice. Further studies are needed to understand the role of PXR signaling in obesity and metabolic disorders in the future. SIGNIFICANCE STATEMENT: The authors demonstrate that deficiency of adipocyte pregnane X receptor (PXR) does not affect diet-induced obesity or metabolic disorders in mice and infers that adipocyte PXR signaling may not play a key role in diet-induced obesity. More studies are needed to understand the tissue-specific role of PXR in obesity.
- Published
- 2023
25. Loss of cAMP Signaling in CD11c Immune Cells Protects Against Diet-Induced Obesity.
- Author
-
Zeng, Liping, Herdman, D Scott, Lee, Sung Min, Tao, Ailin, Das, Manasi, Bertin, Samuel, Eckmann, Lars, Mahata, Sushil K, Wu, Panyisha, Hara, Miki, Byun, Ji-Won, Devulapalli, Shwetha, Patel, Hemal H, Molina, Anthony JA, Osborn, Olivia, Corr, Maripat, Raz, Eyal, and Webster, Nicholas JG
- Subjects
Biomedical and Clinical Sciences ,Immunology ,Obesity ,Diabetes ,Nutrition ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Animals ,Mice ,Adipose Tissue ,White ,Catecholamines ,Diet ,High-Fat ,Insulin ,Insulin Resistance ,Mice ,Inbred C57BL ,Norepinephrine ,Medical and Health Sciences ,Endocrinology & Metabolism ,Biomedical and clinical sciences - Abstract
In obesity, CD11c+ innate immune cells are recruited to adipose tissue and create an inflammatory state that causes both insulin and catecholamine resistance. We found that ablation of Gnas, the gene that encodes Gαs, in CD11c expressing cells protects mice from obesity, glucose intolerance, and insulin resistance. Transplantation studies showed that the lean phenotype was conferred by bone marrow-derived cells and did not require adaptive immunity. Loss of cAMP signaling was associated with increased adipose tissue norepinephrine and cAMP signaling, and prevention of catecholamine resistance. The adipose tissue had reduced expression of catecholamine transport and degradation enzymes, suggesting that the elevated norepinephrine resulted from decreased catabolism. Collectively, our results identified an important role for cAMP signaling in CD11c+ innate immune cells in whole-body metabolism by controlling norepinephrine levels in white adipose tissue, modulating catecholamine-induced lipolysis and increasing thermogenesis, which, together, created a lean phenotype.Article highlightsWe undertook this study to understand how immune cells communicate with adipocytes, specifically, whether cAMP signaling in the immune cell and the adipocyte are connected. We identified a reciprocal interaction between CD11c+ innate immune cells and adipocytes in which high cAMP signaling in the immune cell compartment induces low cAMP signaling in adipocytes and vice versa. This interaction regulates lipolysis in adipocytes and inflammation in immune cells, resulting in either a lean, obesity-resistant, and insulin-sensitive phenotype, or an obese, insulin-resistant phenotype.
- Published
- 2023
26. Fructose induced KHK-C can increase ER stress independent of its effect on lipogenesis to drive liver disease in diet-induced and genetic models of NAFLD
- Author
-
Park, Se-Hyung, Helsley, Robert N, Fadhul, Taghreed, Willoughby, Jennifer LS, Noetzli, Leila, Tu, Ho-Chou, Solheim, Marie H, Fujisaka, Shiho, Pan, Hui, Dreyfuss, Jonathan M, Bons, Joanna, Rose, Jacob, King, Christina D, Schilling, Birgit, Lusis, Aldons J, Pan, Calvin, Gupta, Manoj, Kulkarni, Rohit N, Fitzgerald, Kevin, Kern, Philip A, Divanovic, Senad, Kahn, C Ronald, and Softic, Samir
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Nutrition ,Women's Health ,Prevention ,Genetics ,Digestive Diseases ,Liver Disease ,Chronic Liver Disease and Cirrhosis ,Obesity ,2.1 Biological and endogenous factors ,Oral and gastrointestinal ,Metabolic and endocrine ,Animals ,Female ,Humans ,Male ,Mice ,Diet ,High-Fat ,Fructokinases ,Fructose ,Lipogenesis ,Liver ,Models ,Genetic ,Non-alcoholic Fatty Liver Disease ,Sugar ,Ketohexokinase ,ER stress ,NAFLD ,Endocrinology & Metabolism ,Clinical sciences - Abstract
Non-alcoholic fatty liver disease (NAFLD) is a liver manifestation of metabolic syndrome, and is estimated to affect one billion individuals worldwide. An increased intake of a high-fat diet (HFD) and sugar-sweetened beverages are risk-factors for NAFLD development, but how their combined intake promotes progression to a more severe form of liver injury is unknown. Here we show that fructose metabolism via ketohexokinase (KHK) C isoform leads to unresolved endoplasmic reticulum (ER) stress when coupled with a HFD intake. Conversely, a liver-specific knockdown of KHK in mice consuming fructose on a HFD is adequate to improve the NAFLD activity score and exert a profound effect on the hepatic transcriptome. Overexpression of KHK-C in cultured hepatocytes is sufficient to induce ER stress in fructose free media. Upregulation of KHK-C is also observed in mice with genetically induced obesity or metabolic dysfunction, whereas KHK knockdown in these mice improves metabolic function. Additionally, in over 100 inbred strains of male or female mice hepatic KHK expression correlates positively with adiposity, insulin resistance, and liver triglycerides. Similarly, in 241 human subjects and their controls, hepatic Khk expression is upregulated in early, but not late stages of NAFLD. In summary, we describe a novel role of KHK-C in triggering ER stress, which offers a mechanistic understanding of how the combined intake of fructose and a HFD propagates the development of metabolic complications.
- Published
- 2023
27. Myeloid-specific deletion of chitinase-3-like 1 protein ameliorates murine diet-induced steatohepatitis progression
- Author
-
Kim, Andrea D, Kui, Lin, Kaufmann, Benedikt, Kim, Sung Eun, Leszczynska, Aleksandra, and Feldstein, Ariel E
- Subjects
Medicinal and Biomolecular Chemistry ,Chemical Sciences ,Liver Disease ,Digestive Diseases ,Hepatitis ,Nutrition ,Chronic Liver Disease and Cirrhosis ,Biotechnology ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Inflammatory and immune system ,Oral and gastrointestinal ,Mice ,Humans ,Animals ,Non-alcoholic Fatty Liver Disease ,Chitinases ,Interleukin-13 Receptor alpha2 Subunit ,Liver ,Liver Cirrhosis ,Diet ,High-Fat ,Mice ,Knockout ,Mice ,Inbred C57BL ,Disease Models ,Animal ,Chitinase-like proteins ,Non-alcoholic fatty liver disease ,Hepatic stellate cells ,Infiltrating macrophages ,Immunology ,Medicinal and biomolecular chemistry - Abstract
Chitinase-3-like 1 protein (CHI3L1) is a secreted glycoprotein, strongly correlated with fibrosis severity in chronic liver diseases including non-alcoholic steatohepatitis (NASH). However, the mechanisms by which CHI3L1 contributes to fibrogenesis remain undefined. Here, we showed that infiltrating monocyte-derived liver macrophages represent the main source of CHI3L1 in murine NASH. We developed a floxed CHI3L1 knock-out (KO) mouse to further study the cell-specific role of CHI3L1 ablation. Wildtype (WT) and myeloid cell-specific CHI3L1 KO mice (CreLyz) were challenged with a highly inflammatory and fibrotic dietary model of NASH by administering choline-deficient high-fat diet for 10 weeks. Macrophage accumulation and inflammatory cell recruitment were significantly ameliorated in the CreLyz group compared to WT (F4/80 IHC p
- Published
- 2023
28. Restoration of coronary microvascular function by OGA overexpression in a high-fat diet with low-dose streptozotocin-induced type 2 diabetic mice
- Author
-
Cabrera, Jody Tori, Si, Rui, Tsuji-Hosokawa, Atsumi, Cai, Hua, Yuan, Jason X-J, Dillmann, Wolfgang H, and Makino, Ayako
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Diabetes ,Cardiovascular ,Nutrition ,Heart Disease - Coronary Heart Disease ,Heart Disease ,Aetiology ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Animals ,Mice ,Acetylglucosaminidase ,Diabetes Mellitus ,Experimental ,Diabetes Mellitus ,Type 2 ,Diet ,High-Fat ,Endothelial Cells ,Streptozocin ,cardiovascular disease ,endothelial dysfunction ,angiogenesis ,tube formation ,Pharmacology and Pharmaceutical Sciences ,Medical Physiology ,Endocrinology & Metabolism ,Clinical sciences - Abstract
Sustained hyperglycemia results in excess protein O-GlcNAcylation, leading to vascular complications in diabetes. This study aims to investigate the role of O-GlcNAcylation in the progression of coronary microvascular disease (CMD) in inducible type 2 diabetic (T2D) mice generated by a high-fat diet with a single injection of low-dose streptozotocin. Inducible T2D mice exhibited an increase in protein O-GlcNAcylation in cardiac endothelial cells (CECs) and decreases in coronary flow velocity reserve (CFVR, an indicator of coronary microvascular function) and capillary density accompanied by increased endothelial apoptosis in the heart. Endothelial-specific O-GlcNAcase (OGA) overexpression significantly lowered protein O-GlcNAcylation in CECs, increased CFVR and capillary density, and decreased endothelial apoptosis in T2D mice. OGA overexpression also improved cardiac contractility in T2D mice. OGA gene transduction augmented angiogenic capacity in high-glucose treated CECs. PCR array analysis revealed that seven out of 92 genes show significant differences among control, T2D, and T2D + OGA mice, and Sp1 might be a great target for future study, the level of which was significantly increased by OGA in T2D mice. Our data suggest that reducing protein O-GlcNAcylation in CECs has a beneficial effect on coronary microvascular function, and OGA is a promising therapeutic target for CMD in diabetic patients.
- Published
- 2023
29. Sexual dimorphism in obesity is governed by RELMα regulation of adipose macrophages and eosinophils.
- Author
-
Li, Jiang, Ruggiero-Ruff, Rebecca E, He, Yuxin, Qiu, Xinru, Lainez, Nancy, Villa, Pedro, Godzik, Adam, Coss, Djurdjica, and Nair, Meera G
- Subjects
Adipose Tissue ,Eosinophils ,Macrophages ,Animals ,Mice ,Inbred C57BL ,Mice ,Obesity ,Inflammation ,Sex Characteristics ,Female ,Male ,Diet ,High-Fat ,RELMα ,adipose ,eosinophil ,immunology ,inflammation ,macrophage ,mouse ,obesity ,sexual dimorphism ,Nutrition ,1.1 Normal biological development and functioning ,Aetiology ,2.1 Biological and endogenous factors ,Underpinning research ,Cardiovascular ,Cancer ,Metabolic and endocrine ,Biochemistry and Cell Biology - Abstract
Obesity incidence is increasing worldwide with the urgent need to identify new therapeutics. Sex differences in immune cell activation drive obesity-mediated pathologies where males are more susceptible to obesity comorbidities and exacerbated inflammation. Here, we demonstrate that the macrophage-secreted protein RELMα critically protects females against high-fat diet (HFD)-induced obesity. Compared to male mice, serum RELMα levels were higher in both control and HFD-fed females and correlated with frequency of adipose macrophages and eosinophils. RELMα-deficient females gained more weight and had proinflammatory macrophage accumulation and eosinophil loss in the adipose stromal vascular fraction (SVF), while RELMα treatment or eosinophil transfer rescued this phenotype. Single-cell RNA-sequencing of the adipose SVF was performed and identified sex and RELMα-dependent changes. Genes involved in oxygen sensing and iron homeostasis, including hemoglobin and lncRNA Gm47283/Gm21887, correlated with increased obesity, while eosinophil chemotaxis and response to amyloid-beta were protective. Monocyte-to-macrophage transition was also dysregulated in RELMα-deficient animals. Collectively, these studies implicate a RELMα-macrophage-eosinophil axis in sex-specific protection against obesity and uncover new therapeutic targets for obesity.
- Published
- 2023
30. MBOAT7-driven lysophosphatidylinositol acylation in adipocytes contributes to systemic glucose homeostasis
- Author
-
Massey, William J, Varadharajan, Venkateshwari, Banerjee, Rakhee, Brown, Amanda L, Horak, Anthony J, Hohe, Rachel C, Jung, Bryan M, Qiu, Yunguang, Chan, E Ricky, Pan, Calvin, Zhang, Renliang, Allende, Daniela S, Willard, Belinda, Cheng, Feixiong, Lusis, Aldons J, and Brown, J Mark
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Obesity ,Diabetes ,Digestive Diseases ,Nutrition ,Liver Disease ,Genetics ,Chronic Liver Disease and Cirrhosis ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Oral and gastrointestinal ,Animals ,Mice ,Acylation ,Adipocytes ,Arachidonic Acid ,Diet ,High-Fat ,Glucose ,Homeostasis ,Hyperinsulinism ,Insulin Resistance ,Mice ,Inbred C57BL ,Mice ,Knockout ,Non-alcoholic Fatty Liver Disease ,acyltransferase ,arachidonic acid ,diabetes ,hepatocytes ,hyperinsulinemia ,metabolism ,non-alcoholic fatty liver disease ,obesity ,phosphatidylinositol biosynthesis ,systemic insulin resistance ,Medical Biochemistry and Metabolomics ,Biochemistry & Molecular Biology ,Biochemistry and cell biology ,Medical biochemistry and metabolomics - Abstract
We previously demonstrated that antisense oligonucleotide-mediated knockdown of Mboat7, the gene encoding membrane bound O-acyltransferase 7, in the liver and adipose tissue of mice promoted high fat diet-induced hepatic steatosis, hyperinsulinemia, and systemic insulin resistance. Thereafter, other groups showed that hepatocyte-specific genetic deletion of Mboat7 promoted striking fatty liver and NAFLD progression in mice but does not alter insulin sensitivity, suggesting the potential for cell autonomous roles. Here, we show that MBOAT7 function in adipocytes contributes to diet-induced metabolic disturbances including hyperinsulinemia and systemic insulin resistance. We generated Mboat7 floxed mice and created hepatocyte- and adipocyte-specific Mboat7 knockout mice using Cre-recombinase mice under the control of the albumin and adiponectin promoter, respectively. Here, we show that MBOAT7 function in adipocytes contributes to diet-induced metabolic disturbances including hyperinsulinemia and systemic insulin resistance. The expression of Mboat7 in white adipose tissue closely correlates with diet-induced obesity across a panel of ∼100 inbred strains of mice fed a high fat/high sucrose diet. Moreover, we found that adipocyte-specific genetic deletion of Mboat7 is sufficient to promote hyperinsulinemia, systemic insulin resistance, and mild fatty liver. Unlike in the liver, where Mboat7 plays a relatively minor role in maintaining arachidonic acid-containing PI pools, Mboat7 is the major source of arachidonic acid-containing PI pools in adipose tissue. Our data demonstrate that MBOAT7 is a critical regulator of adipose tissue PI homeostasis, and adipocyte MBOAT7-driven PI biosynthesis is closely linked to hyperinsulinemia and insulin resistance in mice.
- Published
- 2023
31. TGFBI remodels adipose metabolism by regulating the Notch-1 signaling pathway.
- Author
-
Kim, In-San, Park, Hee-Sae, Kwon, Taeg, Nam, Ju-Ock, Lee, Seul, Chae, Jongbeom, Woo, Seon, Seo, Seung, Kim, Ha-Jeong, Kim, Sang-Yeob, and Schlaepfer, David
- Subjects
Mice ,Animals ,Transforming Growth Factor beta ,Obesity ,Adipose Tissue ,Adipocytes ,Signal Transduction ,Diet ,High-Fat ,Mice ,Inbred C57BL ,Insulin Resistance ,Adipose Tissue ,White - Abstract
Extracellular matrix proteins are associated with metabolically healthy adipose tissue and regulate inflammation, fibrosis, angiogenesis, and subsequent metabolic deterioration. In this study, we demonstrated that transforming growth factor-beta (TGFBI), an extracellular matrix (ECM) component, plays an important role in adipose metabolism and browning during high-fat diet-induced obesity. TGFBI KO mice were resistant to adipose tissue hypertrophy, liver steatosis, and insulin resistance. Furthermore, adipose tissue from TGFBI KO mice contained a large population of CD11b+ and CD206+ M2 macrophages, which possibly control adipokine secretion through paracrine mechanisms. Mechanistically, we showed that inhibiting TGFBI-stimulated release of adipsin by Notch-1-dependent signaling resulted in adipocyte browning. TGFBI was physiologically bound to Notch-1 and stimulated its activation in adipocytes. Our findings revealed a novel protective effect of TGFBI deficiency in obesity that is realized via the activation of the Notch-1 signaling pathway.
- Published
- 2023
32. Oat protein modulates cholesterol metabolism and improves cardiac systolic function in high fat, high sucrose fed rats.
- Author
-
Bouchard, Jenny, Raj, Pema, Yu, Liping, Sobhi, Babak, Malalgoda, Maneka, Malunga, Lovemore, Netticadan, Thomas, and Joseph Thandapilly, Sijo
- Subjects
- *
HEART anatomy , *HEART physiology , *CHOLESTEROL metabolism , *OATS , *BIOLOGICAL models , *DIETARY sucrose , *ANTILIPEMIC agents , *RESEARCH funding , *CASEINS , *CARDIOTONIC agents , *GLUCOSE tolerance tests , *DIETARY fats , *HEART , *OXIDATIVE stress , *LDL cholesterol , *RATS , *ANIMAL experimentation , *CHOLESTEROL , *METABOLIC syndrome , *CARDIAC contraction , *INFLAMMATION , *LIVER , *PLANT proteins , *ECHOCARDIOGRAPHY , *BLOOD pressure measurement , *KIDNEYS - Abstract
Oats are recognized to provide many health benefits that are mainly associated with its dietary fibre, β-glucan. However, the protein derived from oats is largely understudied with respect to its ability to maintain health and attenuate risk factors of chronic diseases. The goal of the current study was to investigate the metabolic effects of oat protein consumption in lieu of casein as the protein source in high fat, high sucrose (HF/HS) fed Wistar rats. Four-week-old rats were divided into three groups and were fed three different experimental diets: a control diet with casein as the protein source, an HF/HS diet with casein, or an HF/HS diet with oat protein for 16 weeks. Heart structure and function were determined by echocardiography. Blood pressure measurements, an oral glucose tolerance test, and markers of cholesterol metabolism, oxidative stress, inflammation, and liver and kidney damage were also performed. Our study results show that incorporation of oat protein in the diet was effective in preserving systolic heart function in HF/HS fed rats. Oat protein significantly reduced serum total and low-density lipoprotein cholesterol levels. Furthermore, oat protein normalized liver HMG-CoAR activity, which, to our knowledge, is the first time this has been reported in the literature. Therefore, our research suggests that oat protein can provide hypocholesterolemic and cardioprotective benefits in a diet-induced model of metabolic syndrome. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. Efficacy and safety of ketogenic diet in infants with epilepsy: KIWE RCT
- Author
-
Natasha E Schoeler, Louise Marston, Laura Lyons, Sally Halsall, Ruchika Jain, Siobhan Titre-Johnson, Maryam Balogun, Simon J R Heales, Simon Eaton, Michael Orford, Elizabeth Neal, Christin Eltze, Elma Stephen, Andrew A Mallick, Finbar O’Callaghan, Shakti Agrawal, Alasdair Parker, Martin Kirkpatrick, Andreas Brunklaus, Ailsa McLellan, Helen McCullagh, Rajib Samanta, Rachel Kneen, Hui Jeen Tan, Anita Devlin, Manish Prasad, Rohini Rattihalli, Helen Basu, Archana Desurkar, Ruth Williams, Penny Fallon, Irwin Nazareth, Nicholas Freemantle, and J Helen Cross
- Subjects
high-fat ,low-carbohydrate ,seizures ,randomised controlled trial ,infants ,Medicine - Abstract
Background Many infancy-onset epilepsies have a poor prognosis for seizure control and neurodevelopmental outcome. Ketogenic diets can improve seizures in older children and adults unresponsive to antiseizure medicines. We aimed to determine the effectiveness of the ketogenic diet in reducing seizure frequency compared to further antiseizure medicine in infants with drug-resistant epilepsy. Methods In this randomised, open-label trial, 136 infants with epilepsy, aged 1–24 months, with > 4 seizures/week and a previous trial of ≥ 2 antiseizure medicines were recruited from 19 hospitals in the United Kingdom. Following a 1- or 2-week observation period, participants were randomised to receive the classical ketogenic diet or a further antiseizure medicine for 8 weeks, using a computer-generated schedule without stratification. Treatment allocation was concealed from research nurses involved in patient care, but not from participants. The primary outcome was the number of seizures/day recorded during weeks 6–8. All analyses were intention to treat. The trial is registered with the European Union Drug Regulating Authorities Clinical Trials Database (2013-002195-40). Findings Between 1 January 2015 and 30 September 2021, 136 eligible infants were randomised. Sixty-one (78%) of 78 assigned to a ketogenic diet and 47 (81%) of 58 assigned to antiseizure medicine had primary outcome data. At 8 weeks, the number of seizures per day, accounting for the baseline rate and randomised group, was not significantly different between groups [median (interquartile range) ketogenic diet 5 (1, 16); antiseizure medicine 3 (2, 11), incidence rate ratio 1.33, 95%, confidence internal 0.84 to 2.11; p = 0.22]. A similar number of infants reported at least one serious adverse event in both groups [antiseizure medicine: 24/56 (43%), ketogenic diet: 40/78 (51%)]. The most common serious adverse events were seizures in both groups. Three infants died during the course of the trial, all of whom were randomised to the ketogenic diet arm; deaths were considered to be unrelated to treatment. Interpretation There was no evidence that a ketogenic diet was better than further antiseizure medicine in achieving seizure control in infants with epilepsy. The two treatments were similarly tolerated and a ketogenic diet appears safe to use in infants with epilepsy. A ketogenic diet could be a treatment option in infants whose seizures continue despite trial of two standard antiseizure medicines. Study registration This study was registered as EudraCT 2013-002195-40. Funding This award was funded by the National Institute for Health and Care Research (NIHR) Efficacy and Mechanism Evaluation (EME) programme (NIHR award ref: 12/10/18) and is published in full in Efficacy and Mechanism Evaluation; Vol. 11, No. 16. See the NIHR Funding and Awards website for further award information. Plain language summary Many babies with epilepsy continue to have seizures (‘fits’), despite taking medicines. These babies are unlikely to make the same developmental progress as babies without epilepsy. Ketogenic diets are high in fat and low in carbohydrate and have been shown to reduce seizures in older children and adults. Babies (age 1–24 months) with epilepsy, who still had seizures despite having tried medicines, either started a ketogenic diet or another medicine. They then stayed on this treatment for 8 weeks. We wanted to know whether there was a difference in the number of seizures in the last 2 weeks of treatments compared to before. Seventy-eight babies started a ketogenic diet and 58 started another medicine. Overall, the number of seizures babies had every day was similar in both groups. A similar number of babies in both groups either had to go to hospital for some reason or stay in hospital for longer. This was mostly because of seizures. Two babies suffered ‘sudden unexpected death in epilepsy’ and another died during a planned surgery. These deaths were not thought to be related to the ketogenic diet. Both the ketogenic diet and the medicines helped to reduce the number of seizures in the babies. Babies in both groups had similar side effects. Doctors could consider starting a ketogenic diet in babies who continue to have seizures even though they have tried medicines to stop their seizures. Scientific summary Background Many infancy-onset epilepsies are poorly responsive to antiseizure medicines (ASMs) with poor prognosis for neurodevelopmental outcome. Ketogenic diets (KDs), which are high-fat, low-carbohydrate diets, have been shown to reduce seizures in older children with drug-resistant epilepsy. No high-quality evidence is available for infants. Objectives In this open-label randomised controlled trial, we compared the efficacy of the classical KD to a further appropriate ASM in infants with drug-resistant epilepsy. The primary outcome was the number of seizures recorded during weeks 6–8, accounting for the baseline rate and randomised group. Secondary outcomes at 8 weeks were the number of infants seizure-free in weeks 6–8 of the intervention period, responder rate (defined as the number showing more than a 50% improvement in seizure frequency compared to baseline), tolerance to KD and relationship between medium-chain fatty acids and seizure control. Secondary outcomes at 12 months were retention on treatment, quality of life and neurodevelopmental outcome. Adverse events were recorded throughout the trial. Serious adverse events were reported to the study sponsor. Methods Infants (age 1–24 months) with epilepsy, with an average ≥ 4 seizures/week and previous trials of ≥ 2 ASMs were recruited from 19 hospitals in the UK. Following a 1- or 2-week observation period, during which there were no changes to regular ASMs (emergency seizure treatments continued as required), participants were randomised to receive a classical KD or a further ASM for 8 weeks, using a computer-generated schedule without stratification. An allocation ratio of 1 : 1.35 was used to account for the therapist effect in the KD group only. Treatment allocation was concealed from research nurses involved in patient care, but not from participants. The primary outcome was the number of seizures/day recorded during weeks 6–8. All analyses were intention to treat. The following assessments were performed in all infants prior to randomisation: medical history, physical examination, administration of the Infant Toddler Quality of Life Questionnaire™ (ITQOL-97; © HealthActCHQ Inc. 2013) and Vineland Adaptive Behaviour Scales (Vineland™-II), and clinical laboratory assessments. Food diaries required for diet calculation were returned from parents/guardians of all participants a maximum of 1 week into the observation period. Parents/carers were asked to keep daily seizure diaries throughout the 8-week treatment periods for participants in both arms. Thereafter, they were requested to reduce seizure recording to at least 1–2 days per week, as clinically indicated, until 28 days before the final 12-month visit, when daily seizure recording recommenced. Follow-up visits were arranged at 4 weeks, 8 weeks, and 6, 9 and 12 months. Assessments included clinical review, physical examination, documentation of seizure frequency from seizure diaries, review of adverse events and concomitant medication, clinical laboratory assessments (8 weeks, and 6 and 12 months) and completion of tolerability questionnaire by parents/carers together with research nurses, the ITQOL-97 (8 weeks and 12 months) and Vineland-II (12 months). After the 8-week assessment, according to the infant’s clinical response to treatment (seizure outcome and tolerability), KD or ASM was then continued or changed; those randomised to the ASM arm then had the opportunity to start KD. Results Of 136 eligible infants, 78 were randomised to KD and 58 to ASM. Of 78 infants who started KD, 67 (86%) continued to 8 weeks, of which 61 (78%) had primary outcome data available; 53 (91% of those randomised to ASM group) started a further ASM, 49 (84%) continued to 8 weeks and 47 (81%) had primary outcome data available. The median number of daily seizures was not significantly different in both groups at 8 weeks [KD 5 (1, 16); ASM 3 (2, 11), incidence rate ratio (IRR) 1.33, 95% confidence interval (CI) 0.84 to 2.11; p = 0.22]. The odds ratio (OR) of achieving ≥ 50% seizure reduction was 1.21 (95% CI 0.55 to 2.65) and 0.88 (0.27 to 2.80) for seizure freedom. A total of 7/63 infants (11%) in the KD group were seizure-free, compared with 6/48 (13%) in the ASM group (OR 0.88, 95% CI 0.27 to 2.80). A higher proportion of infants in the ASM group changed the number or dose of concurrent ASMs during the intervention period [24/48 (50%)] compared to KD [9/66 (14%)]. The side-effect score at 8 weeks was similar in both groups [KD median 40; interquartile range (IQR) 38–42; ASM median 41 IQR 39‒44) and there were no clinically significant differences other than those expected in clinical or laboratory parameters between groups. At 8 weeks, median scores within the ITQOL-97 were numerically higher (suggesting better health) in the KD group for 7 of the 12 concepts. The infant’s pain, its global behaviour, impact on parental time and family cohesion were equal between the two groups, although general perceptions of the infant’s health were numerically higher in the ASM arm. A numerically larger proportion of parents/guardians of infants in the KD group perceived their child’s health to be ‘much better than a year ago’ (10/40, 25%) compared to those in the ASM group (3/32, 9%); numerically more parents/guardians of infants in the ASM group perceived their child’s health to be ‘much worse than a year ago’ (8/32, 25%) compared to those in the KD group (2/40, 5%). Of 66 infants randomised to KD > 12 months before the study end date, 31 (47%) continued the diet to 12 months; of 47 randomised to further ASM > 12 months before the study end date, 21 (45%) continued the ASM to 12 months. For those who reported data, there were no differences between groups for any concept within the ITQOL-97 at 12 months, except for the infant’s temperament and mood (coefficient −6.09, 95% CI −11.63 to −0.54) and the infant getting along with others (coefficient −6.79, 95% CI −12.97 to −0.60), which favoured the ASM group. A similar proportion of parents/guardians of infants in both groups perceived their child’s health to be ‘much better than a year ago’ (12/24 50% ASM; 11/30 37% KD) or ‘much worse than a year ago’ (0/24, 0% ASM; 1/30, 3% KD). Within the Vineland-II there were neither significant differences between groups in the overall standardised score nor domain standard scores at 12 months. The Daily living domain sum of v-scale scores was nominally improved in the ASM group (coefficient 2.23, 95% CI −4.22 to −0.25). A total of 73 serious adverse events (SAEs) were reported in the ASM group and 161 in the KD group. A similar proportion of infants in both groups reported at least 1 SAE (43% ASM; 51% KD) – most commonly seizures. Three infants died in the KD arm, all considered unrelated to treatment. Conclusions There was no evidence that KD was better than further ASM in achieving seizure control in infants with epilepsy. The two treatments were similarly tolerated and KD appeared safe to use in infants with epilepsy. KD could be a treatment option in infants whose seizures continue despite trial of two standard ASMs. Further trials are needed with larger cohorts at 12-month follow-up and beyond, particularly to look at quality of life and neurodevelopment, perhaps with alternative study design. Study registration This study was registered as EudraCT 2013-002195-40. Funding This award was funded by the National Institute for Health and Care Research (NIHR) Efficacy and Mechanism Evaluation (EME) programme (NIHR award ref: 12/10/18) and is published in full in Efficacy and Mechanism Evaluation; Vol. 11, No. 16. See the NIHR Funding and Awards website for further award information.
- Published
- 2024
- Full Text
- View/download PDF
34. Insulin-regulated serine and lipid metabolism drive peripheral neuropathy
- Author
-
Handzlik, Michal K, Gengatharan, Jivani M, Frizzi, Katie E, McGregor, Grace H, Martino, Cameron, Rahman, Gibraan, Gonzalez, Antonio, Moreno, Ana M, Green, Courtney R, Guernsey, Lucie S, Lin, Terry, Tseng, Patrick, Ideguchi, Yoichiro, Fallon, Regis J, Chaix, Amandine, Panda, Satchidananda, Mali, Prashant, Wallace, Martina, Knight, Rob, Gantner, Marin L, Calcutt, Nigel A, and Metallo, Christian M
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Prevention ,Neurosciences ,Neurodegenerative ,Nutrition ,Diabetes ,Peripheral Neuropathy ,2.1 Biological and endogenous factors ,5.1 Pharmaceuticals ,Metabolic and endocrine ,Animals ,Mice ,Diabetes Mellitus ,Experimental ,Glycine ,Insulin ,Lipid Metabolism ,Peripheral Nervous System Diseases ,Serine ,Diet ,High-Fat ,Adiposity ,Sphingolipids ,Small Fiber Neuropathy ,Dyslipidemias ,General Science & Technology - Abstract
Diabetes represents a spectrum of disease in which metabolic dysfunction damages multiple organ systems including liver, kidneys and peripheral nerves1,2. Although the onset and progression of these co-morbidities are linked with insulin resistance, hyperglycaemia and dyslipidaemia3-7, aberrant non-essential amino acid (NEAA) metabolism also contributes to the pathogenesis of diabetes8-10. Serine and glycine are closely related NEAAs whose levels are consistently reduced in patients with metabolic syndrome10-14, but the mechanistic drivers and downstream consequences of this metabotype remain unclear. Low systemic serine and glycine are also emerging as a hallmark of macular and peripheral nerve disorders, correlating with impaired visual acuity and peripheral neuropathy15,16. Here we demonstrate that aberrant serine homeostasis drives serine and glycine deficiencies in diabetic mice, which can be diagnosed with a serine tolerance test that quantifies serine uptake and disposal. Mimicking these metabolic alterations in young mice by dietary serine or glycine restriction together with high fat intake markedly accelerates the onset of small fibre neuropathy while reducing adiposity. Normalization of serine by dietary supplementation and mitigation of dyslipidaemia with myriocin both alleviate neuropathy in diabetic mice, linking serine-associated peripheral neuropathy to sphingolipid metabolism. These findings identify systemic serine deficiency and dyslipidaemia as novel risk factors for peripheral neuropathy that may be exploited therapeutically.
- Published
- 2023
35. FGF21 protects against hepatic lipotoxicity and macrophage activation to attenuate fibrogenesis in nonalcoholic steatohepatitis.
- Author
-
Liu, Cong, Schönke, Milena, Spoorenberg, Borah, Lambooij, Joost, van der Zande, Hendrik, Zhou, Enchen, Tushuizen, Maarten, Andreasson, Anne-Christine, Park, Andrew, Oldham, Stephanie, Uhrbom, Martin, Ahlstedt, Ingela, Ikeda, Yasuhiro, Wallenius, Kristina, Peng, Xiao-Rong, Guigas, Bruno, Boon, Mariëtte, Wang, Yanan, and Rensen, Patrick
- Subjects
fibroblast growth factor 21 ,immunology ,inflammation ,lipid/scar-associated macrophages ,liver-adipose tissue crosstalk ,medicine ,mouse ,steatohepatitis ,Mice ,Humans ,Animals ,Non-alcoholic Fatty Liver Disease ,Diabetes Mellitus ,Type 2 ,Macrophage Activation ,Cicatrix ,Liver ,Inflammation ,Diet ,High-Fat ,Cholesterol ,Lipids ,Mice ,Inbred C57BL ,Disease Models ,Animal - Abstract
Analogues of the hepatokine fibroblast growth factor 21 (FGF21) are in clinical development for type 2 diabetes and nonalcoholic steatohepatitis (NASH) treatment. Although their glucose-lowering and insulin-sensitizing effects have been largely unraveled, the mechanisms by which they alleviate liver injury have only been scarcely addressed. Here, we aimed to unveil the mechanisms underlying the protective effects of FGF21 on NASH using APOE*3-Leiden.CETP mice, a well-established model for human-like metabolic diseases. Liver-specific FGF21 overexpression was achieved in mice, followed by administration of a high-fat high-cholesterol diet for 23 weeks. FGF21 prevented hepatic lipotoxicity, accompanied by activation of thermogenic tissues and attenuation of adipose tissue inflammation, improvement of hyperglycemia and hypertriglyceridemia, and upregulation of hepatic programs involved in fatty acid oxidation and cholesterol removal. Furthermore, FGF21 inhibited hepatic inflammation, as evidenced by reduced Kupffer cell (KC) activation, diminished monocyte infiltration, and lowered accumulation of monocyte-derived macrophages. Moreover, FGF21 decreased lipid- and scar-associated macrophages, which correlated with less hepatic fibrosis as demonstrated by reduced collagen accumulation. Collectively, hepatic FGF21 overexpression limits hepatic lipotoxicity, inflammation, and fibrogenesis. Mechanistically, FGF21 blocks hepatic lipid influx and accumulation through combined endocrine and autocrine signaling, respectively, which prevents KC activation and lowers the presence of lipid- and scar-associated macrophages to inhibit fibrogenesis.
- Published
- 2023
36. Impact of various high fat diets on gene expression and the microbiome across the mouse intestines
- Author
-
Martinez-Lomeli, Jose, Deol, Poonamjot, Deans, Jonathan R, Jiang, Tao, Ruegger, Paul, Borneman, James, and Sladek, Frances M
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Cancer ,Autoimmune Disease ,Inflammatory Bowel Disease ,Digestive Diseases ,Colo-Rectal Cancer ,Human Genome ,Biotechnology ,Cancer Genomics ,Microbiome ,Nutrition ,Liver Disease ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Oral and gastrointestinal ,Animals ,Mice ,Diet ,High-Fat ,Soybean Oil ,Dietary Fats ,Microbiota ,Fatty Acids ,Ileum ,Gene Expression ,Colonic Neoplasms ,Inflammatory Bowel Diseases - Abstract
High fat diets (HFDs) have been linked to several diseases including obesity, diabetes, fatty liver, inflammatory bowel disease (IBD) and colon cancer. In this study, we examined the impact on intestinal gene expression of three isocaloric HFDs that differed only in their fatty acid composition-coconut oil (saturated fats), conventional soybean oil (polyunsaturated fats) and a genetically modified soybean oil (monounsaturated fats). Four functionally distinct segments of the mouse intestinal tract were analyzed using RNA-seq-duodenum, jejunum, terminal ileum and proximal colon. We found considerable dysregulation of genes in multiple tissues with the different diets, including those encoding nuclear receptors and genes involved in xenobiotic and drug metabolism, epithelial barrier function, IBD and colon cancer as well as genes associated with the microbiome and COVID-19. Network analysis shows that genes involved in metabolism tend to be upregulated by the HFDs while genes related to the immune system are downregulated; neurotransmitter signaling was also dysregulated by the HFDs. Genomic sequencing also revealed a microbiome altered by the HFDs. This study highlights the potential impact of different HFDs on gut health with implications for the organism as a whole and will serve as a reference for gene expression along the length of the intestines.
- Published
- 2023
37. Microbiota-Dependent Upregulation of Bitter Taste Receptor Subtypes in the Mouse Large Intestine in High-Fat Diet-Induced Obesity
- Author
-
Caremoli, Filippo, Huynh, Jennifer, Lagishetty, Venu, Markovic, Daniela, Braun, Jonathan, Dong, Tien S, Jacobs, Jonathan P, and Sternini, Catia
- Subjects
Biomedical and Clinical Sciences ,Nutrition and Dietetics ,Clinical Sciences ,Prevention ,Nutrition ,Microbiome ,Digestive Diseases ,Obesity ,2.1 Biological and endogenous factors ,Cardiovascular ,Stroke ,Oral and gastrointestinal ,Male ,Female ,Mice ,Animals ,Diet ,High-Fat ,Taste ,Up-Regulation ,Gastrointestinal Microbiome ,Mice ,Inbred C57BL ,Cecum ,Microbiota ,Dysbiosis ,antibiotics ,dysbiosis ,gut ,microbiome ,enteroendocrine cells ,peptides ,hormones ,Food Sciences ,Clinical sciences ,Nutrition and dietetics ,Public health - Abstract
Bitter taste receptors (Tas2rs in mice) detect bitterness, a warning signal for toxins and poisons, and are expressed in enteroendocrine cells. We tested the hypothesis that Tas2r138 and Tas2r116 mRNAs are modulated by microbiota alterations induced by a long-term high-fat diet (HFD) and antibiotics (ABX) (ampicillin and neomycin) administered in drinking water. Cecum and colon specimens and luminal contents were collected from C57BL/6 female and male mice for qRT-PCR and microbial luminal 16S sequencing. HFD with/without ABX significantly increased body weight and fat mass at 4, 6, and 8 weeks. Tas2r138 and Tas2r116 mRNAs were significantly increased in mice fed HFD for 8 weeks vs. normal diet, and this increase was prevented by ABX. There was a distinct microbiota separation in each experimental group and significant changes in the composition and diversity of microbiome in mice fed a HFD with/without ABX. Tas2r mRNA expression in HFD was associated with several genera, particularly with Akkermansia, a Gram-negative mucus-resident bacterium. These studies indicate that luminal bacterial composition is affected by sex, diet, and ABX and support a microbial dependent upregulation of Tas2rs in HFD-induced obesity, suggesting an adaptive host response to specific diet-induced dysbiosis.
- Published
- 2023
38. Diet high in linoleic acid dysregulates the intestinal endocannabinoid system and increases susceptibility to colitis in Mice.
- Author
-
Deol, Poonamjot, Ruegger, Paul, Logan, Geoffrey D, Shawki, Ali, Li, Jiang, Mitchell, Jonathan D, Yu, Jacqueline, Piamthai, Varadh, Radi, Sarah H, Hasnain, Sana, Borkowski, Kamil, Newman, John W, McCole, Declan F, Nair, Meera G, Hsiao, Ansel, Borneman, James, and Sladek, Frances M
- Subjects
Animals ,Humans ,Mice ,Colitis ,Inflammatory Bowel Diseases ,Soybean Oil ,Linoleic Acid ,Endocannabinoids ,Diet ,High-Fat ,Gastrointestinal Microbiome ,HNF4α ,IBD ,PUFAs ,adherent invasive E.Coli ,epithelial barrier function ,gut microbiome ,metabolomics ,olive oil ,oxylipins ,soybean oil ,Complementary and Integrative Health ,Crohn's Disease ,Inflammatory Bowel Disease ,Prevention ,Nutrition ,Autoimmune Disease ,Digestive Diseases ,Aetiology ,2.1 Biological and endogenous factors ,Oral and gastrointestinal ,adherent invasive ,E .Coli ,HNF4 alpha ,Microbiology - Abstract
Inflammatory bowel disease (IBD) is a multifactorial disease with increasing incidence in the U.S. suggesting that environmental factors, including diet, are involved. It has been suggested that excessive consumption of linoleic acid (LA, C18:2 omega-6), which must be obtained from the diet, may promote the development of IBD in humans. To demonstrate a causal link between LA and IBD, we show that a high fat diet (HFD) based on soybean oil (SO), which is comprised of ~55% LA, increases susceptibility to colitis in several models, including IBD-susceptible IL10 knockout mice. This effect was not observed with low-LA HFDs derived from genetically modified soybean oil or olive oil. The conventional SO HFD causes classical IBD symptoms including immune dysfunction, increased intestinal epithelial barrier permeability, and disruption of the balance of isoforms from the IBD susceptibility gene Hepatocyte Nuclear Factor 4α (HNF4α). The SO HFD causes gut dysbiosis, including increased abundance of an endogenous adherent invasive Escherichia coli (AIEC), which can use LA as a carbon source. Metabolomic analysis shows that in the mouse gut, even in the absence of bacteria, the presence of soybean oil increases levels of LA, oxylipins and prostaglandins. Many compounds in the endocannabinoid system, which are protective against IBD, are decreased by SO both in vivo and in vitro. These results indicate that a high LA diet increases susceptibility to colitis via microbial and host-initiated pathways involving alterations in the balance of bioactive metabolites of omega-6 and omega-3 polyunsaturated fatty acids, as well as HNF4α isoforms.
- Published
- 2023
39. Variation in the Early Life and Adult Intestinal Microbiome of Intra-Uterine Growth Restricted Rat Offspring Exposed to a High Fat and Fructose Diet.
- Author
-
Maggiotto, Liesbeth V, Ghosh, Shubhamoy, Shin, Bo-Chul, Ganguly, Amit, Lagishetty, Venu, Jacobs, Jonathan P, and Devaskar, Sherin U
- Subjects
Animals ,Humans ,Rats ,Fetal Growth Retardation ,RNA ,Ribosomal ,16S ,Diet ,Child ,Female ,Male ,Diet ,High-Fat ,Pediatric Obesity ,Dysbiosis ,Gastrointestinal Microbiome ,IUGR ,Western diet ,gut ,microbiota ,Nutrition ,Aging ,Digestive Diseases ,Perinatal Period - Conditions Originating in Perinatal Period ,Genetics ,Preterm ,Low Birth Weight and Health of the Newborn ,Obesity ,Prevention ,Pediatric ,Infant Mortality ,Aetiology ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Good Health and Well Being ,Food Sciences ,Nutrition and Dietetics - Abstract
Intra-Uterine Growth Restriction (IUGR) is a risk factor for many adult-onset chronic diseases, such as diabetes and obesity. These diseases are associated with intestinal microbiome perturbations (dysbiosis). The establishment of an intestinal microbiome begins in utero and continues postnatally (PN). Hypercaloric diet-induced dysbiosis is a major driver of childhood obesity. We hypothesized that different postnatal diets superimposed on IUGR will alter the postnatal intestinal microbiome. We compared four experimental rat groups: (1) Ad lib fed regular chow diet pre- and postnatally (CON), (2-3) IUGR induced by maternal caloric restriction prenatally followed postnatally (PN) by either (2) the control diet (IUGR-RC) or (3) High-Fat-high-fructose (IUGR-HFhf) diet, and lastly (4) HFhf ad lib pre- and postnatally (HFhf). Fecal samples were collected from dams and male and female rat offspring at postnatal day 2, 21, and adult day 180 for 16S rRNA gene sequencing. Maternal diet induced IUGR led to dysbiosis of the intestinal microbiome at PN21. Postnatal HFhf diet significantly reduced microbial diversity and worsened dysbiosis reflected by an increased Gammaproteobacteria/Clostridia ratio. Dysbiosis arising from a mismatch between IUGR and a postnatal HFhf diet may contribute to increased risk of the IUGR offspring for subsequent detrimental health problems.
- Published
- 2023
40. SAPS3 subunit of protein phosphatase 6 is an AMPK inhibitor and controls metabolic homeostasis upon dietary challenge in male mice
- Author
-
Yang, Ying, Reid, Michael A, Hanse, Eric A, Li, Haiqing, Li, Yuanding, Ruiz, Bryan I, Fan, Qi, and Kong, Mei
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Liver Disease ,Digestive Diseases ,Nutrition ,Obesity ,Genetics ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Cardiovascular ,Metabolic and endocrine ,Animals ,Male ,Mice ,AMP-Activated Protein Kinases ,Diet ,High-Fat ,Homeostasis ,Mice ,Inbred C57BL ,Mice ,Knockout ,Phosphoprotein Phosphatases - Abstract
Inhibition of AMPK is tightly associated with metabolic perturbations upon over nutrition, yet the molecular mechanisms underlying are not clear. Here, we demonstrate the serine/threonine-protein phosphatase 6 regulatory subunit 3, SAPS3, is a negative regulator of AMPK. SAPS3 is induced under high fat diet (HFD) and recruits the PP6 catalytic subunit to deactivate phosphorylated-AMPK, thereby inhibiting AMPK-controlled metabolic pathways. Either whole-body or liver-specific deletion of SAPS3 protects male mice against HFD-induced detrimental consequences and reverses HFD-induced metabolic and transcriptional alterations while loss of SAPS3 has no effects on mice under balanced diets. Furthermore, genetic inhibition of AMPK is sufficient to block the protective phenotype in SAPS3 knockout mice under HFD. Together, our results reveal that SAPS3 is a negative regulator of AMPK and suppression of SAPS3 functions as a guardian when metabolism is perturbed and represents a potential therapeutic strategy to treat metabolic syndromes.
- Published
- 2023
41. Maternal diet disrupts the placenta-brain axis in a sex-specific manner.
- Author
-
Ceasrine, Alexis, Devlin, Benjamin, Bolton, Jessica, Green, Lauren, Jo, Young, Huynh, Carolyn, Patrick, Bailey, Washington, Kamryn, Sanchez, Cristina, Joo, Faith, Campos-Salazar, A, Lockshin, Elana, Kuhn, Cynthia, Murphy, Susan, Bilbo, Staci, and Simmons, Leigh Ann
- Subjects
Pregnancy ,Male ,Female ,Mice ,Animals ,Humans ,Placenta ,Serotonin ,Brain ,Diet ,High-Fat ,Dietary Fats - Abstract
High maternal weight is associated with detrimental outcomes in offspring, including increased susceptibility to neurological disorders such as anxiety, depression and communicative disorders. Despite widespread acknowledgement of sex biases in the development of these disorders, few studies have investigated potential sex-biased mechanisms underlying disorder susceptibility. Here, we show that a maternal high-fat diet causes endotoxin accumulation in fetal tissue, and subsequent perinatal inflammation contributes to sex-specific behavioural outcomes in offspring. In male offspring exposed to a maternal high-fat diet, increased macrophage Toll-like receptor 4 signalling results in excess microglial phagocytosis of serotonin (5-HT) neurons in the developing dorsal raphe nucleus, decreasing 5-HT bioavailability in the fetal and adult brains. Bulk sequencing from a large cohort of matched first-trimester human samples reveals sex-specific transcriptome-wide changes in placental and brain tissue in response to maternal triglyceride accumulation (a proxy for dietary fat content). Further, fetal brain 5-HT levels decrease as placental triglycerides increase in male mice and male human samples. These findings uncover a microglia-dependent mechanism through which maternal diet can impact offspring susceptibility for neuropsychiatric disorder development in a sex-specific manner.
- Published
- 2022
42. The mitochondrial fission protein Drp1 in liver is required to mitigate NASH and prevents the activation of the mitochondrial ISR
- Author
-
Steffen, Janos, Ngo, Jennifer, Wang, Sheng-Ping, Williams, Kevin, Kramer, Henning F, Ho, George, Rodriguez, Carlos, Yekkala, Krishna, Amuzie, Chidozie, Bialecki, Russell, Norquay, Lisa, Nawrocki, Andrea R, Erion, Mark, Pocai, Alessandro, Shirihai, Orian S, and Liesa, Marc
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Liver Disease ,Digestive Diseases ,Prevention ,Nutrition ,Chronic Liver Disease and Cirrhosis ,Aetiology ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Oral and gastrointestinal ,Animals ,Diet ,High-Fat ,Fibrosis ,Inflammation ,Liver ,Mice ,Mice ,Inbred C57BL ,Mitochondria ,Mitochondrial Dynamics ,RNA ,Small Interfering ,Weight Loss ,NASH ,ISR ,Drp1 ,Oma1 ,Atf4 ,Physiology ,Biochemistry and cell biology - Abstract
ObjectiveThe mitochondrial fission protein Drp1 was proposed to promote NAFLD, as inhibition of hepatocyte Drp1 early in life prevents liver steatosis induced by high-fat diet in mice. However, whether Drp1-knockdown in older mice can reverse established NASH is unknown.MethodsN-acetylgalactosamine-siRNA conjugates, an FDA approved method to deliver siRNA selectively to hepatocytes, were used to knockdown hepatocyte-Drp1 in mice (NAG-Drp1si). NASH was induced in C57BL/6NTac mice by Gubra-Amylin-NASH diet (D09100310, 40% fat, 22% fructose and 2% cholesterol) and treatment with NAG-Drp1si was started at week 24 of diet. Circulating transaminases, liver histology, gene expression of fibrosis and inflammation markers, and hydroxyproline synthesis determined NASH severity. Liver NEFA and triglycerides were quantified by GC/MS. Mitochondrial function was determined by respirometry. Western blots of Oma1, Opa1, p-eIf2α, as well as transcriptional analyses of Atf4-regulated genes determined ISR engagement.ResultsNAG-Drp1si treatment decreased body weight and induced liver inflammation in adult healthy mice. Increased hepatic Gdf15 production was the major contributor to body-weight loss caused by NAG-Drp1si treatment, as Gdf15 receptor deletion (Gfral KO) prevented the decrease in food intake and mitigated weight loss. NAG-Drp1si activated the Atf4-controlled integrated stress response (ISR) to increase hepatic Gdf15 expression. NAG-Drp1si in healthy mice caused ER stress and activated the mitochondrial protease Oma1, which are the ER and mitochondrial triggers that activate the Atf4-controlled ISR. Remarkably, induction of NASH was not sufficient to activate Oma1 in liver. However, NAG-Drp1si treatment was sufficient to activate Oma1 in adult mice with NASH, as well as exacerbating NASH-induced ER stress. Consequently, NAG-Drp1si treatment in mice with NASH led to higher ISR activation, exacerbated inflammation, fibrosis and necrosis.ConclusionDrp1 mitigates NASH by decreasing ER stress, preventing Oma1 activation and ISR exacerbation. The elevation in Gdf15 actions induced by NAG-Drp1si might represent an adaptive response decreasing the nutrient load to liver when mitochondria are misfunctional. Our study argues against blocking Drp1 in hepatocytes to combat NASH.
- Published
- 2022
43. High-Caloric Diets in Adolescence Impair Specific GABAergic Subpopulations, Neurogenesis, and Alter Astrocyte Morphology.
- Author
-
Mota, Bárbara, Brás, Ana Rita, Araújo-Andrade, Leonardo, Silva, Ana, Pereira, Pedro A., Madeira, M. Dulce, and Cardoso, Armando
- Subjects
- *
GLIAL fibrillary acidic protein , *BRAIN-derived neurotrophic factor , *REDUCING diets , *ADOLESCENCE , *NEUROPEPTIDE Y , *GABA , *DEVELOPMENTAL neurobiology , *SUCROSE , *CALRETININ - Abstract
We compared the effects of two different high-caloric diets administered to 4-week-old rats for 12 weeks: a diet rich in sugar (30% sucrose) and a cafeteria diet rich in sugar and high-fat foods. We focused on the hippocampus, particularly on the gamma-aminobutyric acid (GABA)ergic system, including the Ca2+-binding proteins parvalbumin (PV), calretinin (CR), calbindin (CB), and the neuropeptides somatostatin (SST) and neuropeptide Y (NPY). We also analyzed the density of cholinergic varicosities, brain-derived neurotrophic factor (BDNF), reelin (RELN), and cyclin-dependent kinase-5 (CDK-5) mRNA levels, and glial fibrillary acidic protein (GFAP) expression. The cafeteria diet reduced PV-positive neurons in the granular layer, hilus, and CA1, as well as NPY-positive neurons in the hilus, without altering other GABAergic populations or overall GABA levels. The high-sugar diet induced a decrease in the number of PV-positive cells in CA3 and an increase in CB-positive cells in the hilus and CA1. No alterations were observed in the cholinergic varicosities. The cafeteria diet also reduced the relative mRNA expression of RELN without significant changes in BDNF and CDK5 levels. The cafeteria diet increased the number but reduced the length of the astrocyte processes. These data highlight the significance of determining the mechanisms mediating the observed effects of these diets and imply that the cognitive impairments previously found might be related to both the neuroinflammation process and the reduction in PV, NPY, and RELN expression in the hippocampal formation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
44. NLRP3基因敲减对高脂高糖饮食诱导的非酒精性脂肪性肝炎 小鼠模型的影响.
- Author
-
黄 倩, 王卓媛, 安梓铭, 辛 鑫, 孙沁梅, 苟小军, 胡义扬, and 冯 琴
- Abstract
Objective To investigate the effect of NOD-like receptor family pyrin domain containing 3(NLRP3) knockdown on a mouse model of nonalcoholic steatohepatitis (NASH) induced by high-fat high-carbohydrate (HFHC) diet. Methods A total of 44 mice were randomly divided into normal diet group (CON group) with 20 mice and HFHC group with 24 mice. At the end of week 14 of modeling,4 mice were randomly selected from the HFHC group for the pre-experiment of adeno-associated virus (AAV) by tail vein injection, and NLRP3 knockdown was verified after 4 weeks. After NLRP3 knockdown was verified at the end of week 18, the remaining 40 mice were given a single tail vein injection of AAV, and then they were divided into CON+NLRP3 knockdown negative control group (CON+NLRP3-NC group), CON+NLRP3 knockdown group (CON+NLRP3-KD group), HFHC+NLRP3- NC group, and HFHC+NLRP3-KD group, with 10 mice in each group. At the end of week 24, the activation of NLRP3 inflammasome was observed; related indicators were measured, including body weight, liver weight, liver index, and glucose metabolism (fasting blood glucose, fasting insulin, and Homeostasis Model Assessment of Insulin Resistance [HOMA-IR] index);the indicators of liver lipid content (liver triglyceride [TG] and oil red O staining), liver inflammation (serum alanine aminotransferase [ALT] activity, HE staining, and inflammation-related genes), and liver fibrosis (Sirius Red staining and fibrosis-related genes) were measured. A one-way analysis of variance was used for comparison of continuous data between multiple groups, and the least significant difference t-test was used for further comparison between two groups. Results Compared with the CON+NLRP3-NC group based on the results of Western Blot, the HFHC+NLRP3-NC group had significant increases in the protein expression levels of NLRP3, pro-Caspase1, Caspase1, ASC, and IL-1β, while the HFHC+NLRP3-KD group had significant reductions in these levels (all P<0.05). The HFHC+NLRP3-NC group showed varying degrees of increase in body weight, liver weight, liver index, and glucose metabolism indicators, while the HFHC+NLRP3-KD group showed significant improvements in these indicators (all P<0.05). As for hepatic fat deposition, compared with the CON+NLRP3-NC group, the HFHC+NLRP3-NC group had a significant increase in liver TG, with a large number of red lipid droplets shown by oil red O staining, and the HFHC+ NLRP3-KD group had significant reductions in liver TG and the number of lipid droplets in the liver (all P<0.01). In terms of liver inflammation, compared with the CON+NLRP3-NC group, the HFHC+NLRP3-NC group had significant increases in serum ALT, NAFLD activity score, and inflammation-related genes, while the HFHC+NLRP3-KD group had significant reductions in these indicators (all P<0.01). As for liver fibrosis, compared with the CON+NLRP3-NC group, the HFHC+NLRP3-NC group had significant increases in collagen fiber area and fibrosis-related genes, and the HFHC+NLRP3-KD group had significant reductions in fibrosis-related genes (all P<0.05) and a tendency of reduction in collagen fiber area (P>0.05). Conclusion NLRP3 knockdown can significantly improve hepatic fat deposition and inflammation in a mouse model of HFHC-induced NASH. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
45. Genetics and diet shape the relationship between islet function and whole body metabolism.
- Author
-
Yau, Belinda, Madsen, Søren, Nelson, Marin E., Cooke, Kristen C., Fritzen, Andreas M., Thorius, Ida H., Stöckli, Jacqueline, James, David E., and Kebede, Melkam A.
- Subjects
- *
HIGH-fat diet , *WESTERN diet , *ISLANDS , *GLUCOSE metabolism , *GENETICS , *KETOGENIC diet , *GLUCOSE intolerance - Abstract
Despite the fact that genes and the environment are known to play a central role in islet function, our knowledge of how these parameters interact to modulate insulin secretory function remains relatively poor. Presently, we performed ex vivo glucose-stimulated insulin secretion and insulin content assays in islets of 213 mice from 13 inbred mouse strains on chow, Western diet (WD), and a high-fat, carbohydrate-free (KETO) diet. Strikingly, among these 13 strains, islets from the commonly used C57BL/6J mouse strain were the least glucose responsive. Using matched metabolic phenotyping data, we performed correlation analyses of isolated islet parameters and found a positive correlation between basal and glucose-stimulated insulin secretion, but no relationship between insulin secretion and insulin content. Using in vivo metabolic measures, we found that glucose tolerance determines the relationship between ex vivo islet insulin secretion and plasma insulin levels. Finally, we showed that islet glucose-stimulated insulin secretion decreased with KETO in almost all strains, concomitant with broader phenotypic changes, such as increased adiposity and glucose intolerance. This is an important finding as it should caution against the application of KETO diet for beta-cell health. Together these data offer key insights into the intersection of diet and genetic background on islet function and whole body glucose metabolism. NEW & NOTEWORTHY: Thirteen strains of mice on chow, Western diet, and high-fat, carbohydrate-free (KETO), correlating whole body phenotypes to ex vivo pancreatic islet functional measurements, were used. The study finds a huge spectrum of functional islet responses and insulin phenotypes across all strains and diets, with the ubiquitous C57Bl/6J mouse exhibiting the lowest secretory response of all strains, highlighting the overall importance of considering genetic background when investigating islet function. Ex vivo basal and stimulated insulin secretion are correlated in the islet, and KETO imparts widescale downregulation of islet insulin secretion. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
46. Dietary factors in circadian rhythm modulation and their impact on metabolic diseases: a state of the science review.
- Author
-
Dalvi, Malvika and Medithi, Srujana
- Subjects
- *
CIRCADIAN rhythms , *METABOLIC disorders , *HIGH-salt diet , *DISEASE progression , *CLOCK genes - Abstract
Biological functions of living things are regulated by the circadian rhythm, a 24-hour cycle. Master clock in the hypothalamus is dependent on light-dark signals, while peripheral clock (present in organs) is cell-independent being synchronized with central clock. Mammals' circadian clocks control various catabolic and anabolic processes in their peripheral tissues. Peripheral organs display tissue-specific differences in the circadian period and phase and exhibit a circadian oscillator. Various diets will affect the peripheral clock in different organ systems. Ketogenic and high-fat diets can alter clock gene expression, while intermittent fasting shows improved circadian rhythm desynchronization. High-sugar diets can cause circadian rhythm-dependent gene amplification involved in hepatic carbohydrate and fat metabolism, leading to dyslipidemia and disturbed metabolism. High-salt diets can increase peripheral clock gene expression. Different diets can affect the peripheral clock rhythm, causing phase shifts, leading to the desynchronization of the central and peripheral clocks subsequently leading to various metabolic diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
47. Evaluation of Anti-Obesity and Antidiabetic Activities of Orostachys japonicus in Cell and Animal Models.
- Author
-
Lamichhane, Ramakanta, Pandeya, Prakash Raj, Lee, Kyung-Hee, Lamichhane, Gopal, Cheon, Jae-Young, Park, Hyo Shin, Tuan, Nguyen Quoc, and Jung, Hyun-Ju
- Subjects
- *
ADIPOGENESIS , *STAINS & staining (Microscopy) , *ANIMAL models in research , *BLOOD lipids , *HYPOGLYCEMIC agents , *ADIPOSE tissues , *LIPIDS , *FAT - Abstract
Orostachys japonicus is a popular traditional medicinal herb used in Asian countries. This study is focused on evaluating its role in lipid and glucose metabolism in cell and animal models to establish the plant as an anti-obesity and antidiabetic herb. A butanol fraction of O. japonicus was used in the study. The lipid production was evaluated by the Oil Red O technique while the expression of adipogenic markers by Western blotting and RT-PCR using 3T3-L1 preadipocyte. The effect on glucose uptake activity was evaluated in C2C12 myoblast cells. The animal study was carried out in C57BL mice to evaluate anti-obesity activity using the high-fat diet model. The evaluation of serum lipid, blood glucose, adipogenic and fibrosis markers in the liver, and fat deposition in the liver and adipose tissue (by histology) of mice was conducted. Butanol fraction of O. japonicus significantly inhibited the lipid production in the 3T3-L1 cells and reduced the expression of PPARγ, C/EBPα, SREBP-1c and aP2. It enhanced glucose uptake in insulin-resistant C2C12 myoblast cells. It reduced body weight, triglycerides, and blood glucose in the obese mice. It significantly inhibited lipid accumulation in the liver and adipose tissue of obese mice along with suppression of expression of adipogenic and fibrosis markers in the liver. In summary, supporting the previous results, this study helped to establish the potent anti-obesity, antidiabetic, and liver-protecting effect of the butanol fraction of O. japonicus. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
48. Glycerate from intestinal fructose metabolism induces islet cell damage and glucose intolerance
- Author
-
Wu, Yanru, Wong, Chi Wut, Chiles, Eric N, Mellinger, Allyson L, Bae, Hosung, Jung, Sunhee, Peterson, Ted, Wang, Jamie, Negrete, Marcos, Huang, Qiang, Wang, Lihua, Jang, Cholsoon, Muddiman, David C, Su, Xiaoyang, Williamson, Ian, and Shen, Xiling
- Subjects
Digestive Diseases ,Autoimmune Disease ,Diabetes ,Nutrition ,Liver Disease ,Aetiology ,2.1 Biological and endogenous factors ,Oral and gastrointestinal ,Metabolic and endocrine ,Blood Glucose ,Diabetes Mellitus ,Type 2 ,Diet ,High-Fat ,Dietary Fats ,Fructose ,Glucose ,Glucose Intolerance ,Humans ,Insulin ,Islets of Langerhans ,diabetes ,dietary fat ,fructose ,glucose intolerance ,glycerate ,insulin ,intestine ,islet cell ,metabolism ,western diet ,Biochemistry and Cell Biology ,Medical Biochemistry and Metabolomics ,Endocrinology & Metabolism - Abstract
Dietary fructose, especially in the context of a high-fat western diet, has been linked to type 2 diabetes. Although the effect of fructose on liver metabolism has been extensively studied, a significant portion of the fructose is first metabolized in the small intestine. Here, we report that dietary fat enhances intestinal fructose metabolism, which releases glycerate into the blood. Chronic high systemic glycerate levels induce glucose intolerance by slowly damaging pancreatic islet cells and reducing islet sizes. Our findings provide a link between dietary fructose and diabetes that is modulated by dietary fat.
- Published
- 2022
49. Organ-Specific Glucose Uptake: Does Sex Matter?
- Author
-
Gandhi, Adithi, Tang, Ryan, Seo, Youngho, and Bhargava, Aditi
- Subjects
Animals ,Mice ,Diabetes Mellitus ,Type 2 ,Fluorodeoxyglucose F18 ,Glucose ,Longitudinal Studies ,Female ,Male ,Diet ,High-Fat ,Metabolic Syndrome ,18F-fluorodeoxyglucose ,PET scan ,brown fat ,female ,heart ,high-fat diet ,male ,sex differences ,skeletal muscle ,young mice ,Diabetes ,Obesity ,Prevention ,Nutrition ,Metabolic and endocrine ,F-18-fluorodeoxyglucose - Abstract
Glucose uptake by peripheral organs is essential for maintaining blood glucose levels within normal range. Impaired glucose uptake is a hallmark of type 2 diabetes (T2D) and metabolic syndrome and is characterized by insulin resistance. Male sex is an independent risk factor for the development of T2D. We tested whether sex and diet are independent variables for differential glucose uptake by various organs. Here, in a longitudinal study, we used 18F-fluorodeoxyglucose (FDG) and positron emission tomography (PET) to determine baseline differences in whole-body glucose uptake in young male and female mice on chow and high-fat diets. We report that sex and diet are important independent variables that account for differential glucose uptake in brown fat, skeletal muscle, liver, heart, kidney, and the stomach, but not the brain, lungs, pancreas, small intestine, or perigonadal adipose. Of the seven organs analyzed, two organs, namely brown fat, and the heart had the highest concentrations of FDG, followed by the brain, kidneys, and skeletal muscle on chow diet. Young female mice had 47% greater FDG uptake in the brown fat compared to male mice, whereas skeletal muscle FDG uptake was 49% greater in male mice. The high-fat diet inhibited FDG uptake in brown fat, skeletal muscle, and the heart, three major organs involved in uptake, whereas brain uptake was enhanced in both sexes. These foundational and groundbreaking findings suggest that mechanisms of glucose homeostasis are context- and organ-dependent and highlight the need to study sex-specific outcomes and mechanisms for diseases such as T2D, obesity, and metabolic syndrome.
- Published
- 2022
50. IL-4 polarized human macrophage exosomes control cardiometabolic inflammation and diabetes in obesity
- Author
-
Phu, Tuan Anh, Ng, Martin, Vu, Ngan K, Bouchareychas, Laura, and Raffai, Robert L
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Diabetes ,Nutrition ,Obesity ,2.1 Biological and endogenous factors ,Aetiology ,Cardiovascular ,Oral and gastrointestinal ,Metabolic and endocrine ,Adipose Tissue ,Animals ,Cardiovascular Diseases ,Diabetes Mellitus ,Diet ,High-Fat ,Exosomes ,Humans ,Inflammation ,Insulin ,Insulin Resistance ,Interleukin-4 ,Macrophages ,Mice ,Mice ,Inbred C57BL ,MicroRNAs ,PPARγ ,adipocyte ,beiging ,cardiometabolic inflammation ,exosomes ,macrophage ,microRNA-33 ,mitochondrial respiration ,obesity ,type II diabetes ,Technology ,Medical and Health Sciences ,Biotechnology ,Genetics ,Clinical sciences ,Medical biotechnology - Abstract
Cardiometabolic disease is an increasing cause of morbidity and death in society. While M1-like macrophages contribute to metabolic inflammation and insulin resistance, those polarized to an M2-like phenotype exert protective properties. Building on our observations reporting M2-like macrophage exosomes in atherosclerosis control, we tested whether they could serve to control inflammation in the liver and adipose tissue of obese mice. In thinking of clinical translation, we studied human THP-1 macrophages exposed to interleukin (IL)-4 as a source of exosomes (THP1-IL4-exo). Our findings show that THP1-IL4-exo polarized primary macrophages to an anti-inflammatory phenotype and reprogramed their energy metabolism by increasing levels of microRNA-21/99a/146b/378a (miR-21/99a/146b/378a) while reducing miR-33. This increased lipophagy, mitochondrial activity, and oxidative phosphorylation (OXPHOS). THP1-IL4-exo exerted a similar regulation of these miRs in cultured 3T3-L1 adipocytes. This enhanced insulin-dependent glucose uptake through increased peroxisome proliferator activated receptor gamma (PPARγ)-driven expression of GLUT4. It also increased levels of UCP1 and OXPHOS activity, which promoted lipophagy, mitochondrial activity, and beiging of 3T3-L1 adipocytes. Intraperitoneal infusions of THP1-IL4-exo into obese wild-type and Ldlr-/- mice fed a Western high-fat diet reduced hematopoiesis and myelopoiesis, and favorably reprogramed inflammatory signaling and metabolism in circulating Ly6Chi monocytes. This also reduced leukocyte numbers and inflammatory activity in the circulation, aorta, adipose tissue, and the liver. Such treatments reduced hepatic steatosis and increased the beiging of white adipose tissue as revealed by increased UCP1 expression and OXPHOS activity that normalized blood insulin levels and improved glucose tolerance. Our findings support THP1-IL4-exo as a therapeutic approach to control cardiometabolic disease and diabetes in obesity.
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.