1
|
Dajnowicz-Brzezik P, Żebrowska E, Maciejczyk M, Zalewska A, Chabowski A. α -lipoic acid supplementation reduces oxidative stress and inflammation in red skeletal muscle of insulin-resistant rats. Biochem Biophys Res Commun 2025; 742:151107. [PMID: 39667068 DOI: 10.1016/j.bbrc.2024.151107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/16/2024] [Accepted: 11/30/2024] [Indexed: 12/14/2024]
Abstract
α -lipoic acid (ALA) is an eight-carbon saturated fatty acid with strong antioxidant activity. Despite previous reports of ALA's protective properties in treating cardiovascular and metabolic diseases (including insulin resistance and diabetes), little is known about the compound's effects on skeletal muscle metabolism. In particular, the effect of ALA on glycooxidative and nitrosative damage in red muscles during insulin resistance is unknown. This study investigated the therapeutic potential of ALA on the antioxidant barrier as well as oxidative, nitrosative and carbonyl stress in the red skeletal muscle of rats with high-fat diet-induced insulin resistance. Male Wistar cmdb/outbred rats were divided into four equal groups: control diet (CTRL), high fat diet (HFD), CTRL + ALA (30 mg/kg body weight for 4 weeks; intragastrically) and HFD + ALA. Enzymatic and nonenzymatic antioxidant systems, protein and lipid glycoxidation, nitrosative stress, and selected inflammatory/apoptosis parameters were assessed using colorimetric, fluorimetric, and immune-enzymatic methods. ALA lowered body weight and glucose metabolism parameters in insulin-resistant rats. ALA not only strengthened enzymatic antioxidant defense (by increasing superoxide dismutase, catalase and glutathione peroxidase activity) but also stimulated the synthesis of non-enzymatic GSH. ALA supplementation inhibited lipid peroxidation (decreased lipid hydroperoxides and malondialdehyde content) and prevented protein oxidation (by lowering advanced oxidation protein products concentration) in red muscle. ALA's multifactorial actions on muscle tissue also included inhibition of inflammation and apoptosis, requiring further research to elucidate its effects in metabolic diseases.
Collapse
Affiliation(s)
- Patrycja Dajnowicz-Brzezik
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C st., 15-222, Bialystok, Poland.
| | - Ewa Żebrowska
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C st., 15-222, Bialystok, Poland
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, Mickiewicza 2C st., 15-222, Bialystok, Poland
| | - Anna Zalewska
- Independent Laboratory of Experimental Dentistry, Medical University of Bialystok, M. Skłodowskiej-Curie 24A st., 15-276, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C st., 15-222, Bialystok, Poland
| |
Collapse
|
2
|
Naidoo K, Khathi A. Investigating the Effects of Gossypetin on Cardiovascular Function in Diet-Induced Pre-Diabetic Male Sprague Dawley Rats. Int J Mol Sci 2024; 25:12105. [PMID: 39596174 PMCID: PMC11594263 DOI: 10.3390/ijms252212105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/07/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
Gossypetin (GTIN) is a naturally occurring flavonoid recognised for its pharmacological properties. This study examined the effects of GTIN on cardiovascular function in a diet-induced pre-diabetic rat model, which has not been previously studied. Pre-diabetes was induced using a high-fat high-carbohydrate (HFHC) diet supplemented with 15% fructose water for 20 weeks. Thereafter, the pre-diabetic animals were sub-divided into five groups (n = 6), where they were either orally treated with GTIN (15 mg/kg) or metformin (MET) (500 mg/kg), both in the presence and absence of dietary intervention for 12 weeks. The results demonstrated that the pre-diabetic (PD) control group exhibited significantly higher plasma triglyceride, total cholesterol, low-density lipoprotein and very low-density lipoprotein levels, along with decreased high-density lipoprotein (HDL) levels in comparison to the non-pre-diabetic (NPD) group. This was accompanied by significantly higher mean arterial pressure (MAP), body mass index (BMI), waist circumference (WC) and plasma endothelial nitric oxide (eNOS) levels in PD control. Additionally, there were increased heart malondialdehyde levels, reduced heart superoxide dismutase and glutathione peroxidase activity as well as increased plasma interleukin-6, tumour necrosis factor alpha and c-reactive protein levels present in the PD control group. Notably, both GTIN-treated groups showed significantly reduced plasma lipid levels and increased HDL, as well as decreases in MAP, BMI, WC and eNOS levels in comparison to PD control. Additionally, GTIN significantly decreased heart lipid peroxidation, enhanced antioxidant activity and decreased plasma inflammation markers. These findings may suggest that GTIN administration in both the presence and absence of dietary intervention may offer therapeutic potential in ameliorating cardiovascular disturbances associated with the PD state. However, future studies are needed to determine the physiological mechanisms by which GTIN improves cardiovascular function in the PD state.
Collapse
Affiliation(s)
| | - Andile Khathi
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa;
| |
Collapse
|
3
|
Wali JA, Ni D, Raubenheimer D, Simpson SJ. Macronutrient interactions and models of obesity: Insights from nutritional geometry. Bioessays 2024:e2400071. [PMID: 39506509 DOI: 10.1002/bies.202400071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/24/2024] [Indexed: 11/08/2024]
Abstract
The global obesity epidemic results from a complex interplay of genetic and environmental factors, with diet being a prominent modifiable element driving weight gain and adiposity. Although excess intake of energetic macronutrients is implicated in causing obesity, ongoing debate centers on whether sugar or fat or both are driving the rising obesity rates. This has led to competing models of obesity such as the "Carbohydrate Insulin Model", the "Energy Balance Model", and the "Fructose Survival Hypothesis". Conflicting evidence from studies designed to focus on individual energetic macronutrients or energy rather than macronutrient mixtures underlies this disagreement. Recent research in humans and animals employing the nutritional geometry framework (NGF) emphasizes the importance of considering interactions among dietary components. Protein interacts with carbohydrates, fats, and dietary energy density to influence both calorie intake ("protein leverage") and, directly and indirectly, metabolic physiology and adiposity. Consideration of these interactions can help to reconcile different models of obesity, and potentially cast new light on obesity interventions.
Collapse
Affiliation(s)
- Jibran A Wali
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Duan Ni
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Chronic Diseases Theme, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - David Raubenheimer
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
4
|
Nkosi A, Pather R, Mshengu B, Khathi A, Ngubane P. Establishing a Female Animal Model of Prediabetes Using a High-Carbohydrate, High-Fat Diet. Curr Issues Mol Biol 2024; 46:12397-12416. [PMID: 39590330 PMCID: PMC11592985 DOI: 10.3390/cimb46110736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
Prediabetes is a condition that often precedes the onset of type 2 diabetes and is characterized by moderate levels of insulin resistance. This condition is well established in male animal models for diabetes; however, few female models exist. There is accumulating evidence that sex variations affect the pathogenesis, treatment, and consequences of numerous diseases, such as type 2 diabetes. Therefore, we sought to develop a diet-induced prediabetic female animal model to better understand prediabetes development and its effects in females. Female Sprague Dawley rats were randomly allocated to one of two groups: the standard diet (SD) group fed a standard diet with normal drinking water, and the high-carbohydrate, high-fat (HCHF) group fed a high-carbohydrate and high-fat diet with drinking water supplemented with fructose. During induction, we measured food intake, body weight, body mass index (BMI), and oral glucose tolerance response (OGT). After the induction period, biochemical analyses were conducted to assess the levels of plasma leptin, ghrelin, insulin, and glycated hemoglobin (HbA1c). Glycogen concentrations were quantified in the liver and skeletal muscles. The HCHF diet-fed group presented higher body weight gain, food intake, and BMI levels, which were accompanied by elevated plasma insulin, ghrelin, and liver and skeletal muscle glycogen levels compared to the SD-fed group. In the HCHF diet-fed group, the HOMA-IR was above 1.9, suggesting the presence of moderate levels of insulin resistance. The OGT response was significantly higher in the HCHF-fed group versus the SD-fed group, suggesting impaired glucose tolerance, thus displaying the signs and symptoms of prediabetes. The HCHF diet with fructose led to the induction of prediabetes in female Sprague Dawley rats. This model could be used to investigate and outline the pathophysiological complications associated with prediabetes in females as a result of the prolonged ingestion of a high carbohydrate, high-fat diet with fructose. The development of this model could also serve as an effort to further bridge the gap regarding the inclusion of females in biomedical research, thus providing advancements in deriving better, specified treatment strategies for women.
Collapse
Affiliation(s)
- Ayanda Nkosi
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Westville, Durban 4000, South Africa; (A.N.); (B.M.); (A.K.)
| | - Reveshni Pather
- Department of Human Physiology, University of KwaZulu-Natal, Westville, Durban 4000, South Africa;
| | - Bongeka Mshengu
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Westville, Durban 4000, South Africa; (A.N.); (B.M.); (A.K.)
| | - Andile Khathi
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Westville, Durban 4000, South Africa; (A.N.); (B.M.); (A.K.)
| | - Phikelelani Ngubane
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Westville, Durban 4000, South Africa; (A.N.); (B.M.); (A.K.)
| |
Collapse
|
5
|
Ansari P, Khan JT, Chowdhury S, Reberio AD, Kumar S, Seidel V, Abdel-Wahab YHA, Flatt PR. Plant-Based Diets and Phytochemicals in the Management of Diabetes Mellitus and Prevention of Its Complications: A Review. Nutrients 2024; 16:3709. [PMID: 39519546 PMCID: PMC11547802 DOI: 10.3390/nu16213709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/27/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Diabetes mellitus (DM) is currently regarded as a global public health crisis for which lifelong treatment with conventional drugs presents limitations in terms of side effects, accessibility, and cost. Type 2 diabetes (T2DM), usually associated with obesity, is characterized by elevated blood glucose levels, hyperlipidemia, chronic inflammation, impaired β-cell function, and insulin resistance. If left untreated or when poorly controlled, DM increases the risk of vascular complications such as hypertension, nephropathy, neuropathy, and retinopathy, which can be severely debilitating or life-threatening. Plant-based foods represent a promising natural approach for the management of T2DM due to the vast array of phytochemicals they contain. Numerous epidemiological studies have highlighted the importance of a diet rich in plant-based foods (vegetables, fruits, spices, and condiments) in the prevention and management of DM. Unlike conventional medications, such natural products are widely accessible, affordable, and generally free from adverse effects. Integrating plant-derived foods into the daily diet not only helps control the hyperglycemia observed in DM but also supports weight management in obese individuals and has broad health benefits. In this review, we provide an overview of the pathogenesis and current therapeutic management of DM, with a particular focus on the promising potential of plant-based foods.
Collapse
Affiliation(s)
- Prawej Ansari
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama, Birmingham (UAB), Birmingham, AL 35233, USA
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
- Centre for Diabetes Research, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (Y.H.A.A.-W.); (P.R.F.)
| | - Joyeeta T. Khan
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR 72205, USA
| | - Suraiya Chowdhury
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
| | - Alexa D. Reberio
- School of Pharmacy and Public Health, Department of Pharmacy, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
| | - Sandeep Kumar
- Comprehensive Diabetes Center, Heersink School of Medicine, University of Alabama, Birmingham (UAB), Birmingham, AL 35233, USA
| | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK;
| | - Yasser H. A. Abdel-Wahab
- Centre for Diabetes Research, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (Y.H.A.A.-W.); (P.R.F.)
| | - Peter R. Flatt
- Centre for Diabetes Research, School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (Y.H.A.A.-W.); (P.R.F.)
| |
Collapse
|
6
|
Ubaldo-Reyes LM, Espitia-Bautista E, Barajas-Martínez A, Martínez-Tapia R, Rodríguez-Mata V, Noriega-Navarro R, Escalona R, Castillo-Hernández J, Pérez-Torres A, Navarro L. High-Fat Diet-Induced Blood-Brain Barrier Dysfunction: Impact on Allodynia and Motor Coordination in Rats. Int J Mol Sci 2024; 25:11218. [PMID: 39457000 PMCID: PMC11508281 DOI: 10.3390/ijms252011218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
The associations among increased pain sensitivity, obesity, and systemic inflammation have not been described as related to BBB dysfunctions. To analyze the metabolic, behavioral, and inflammatory effects of a high-fat diet (HFD) and ultrastructural modifications in brain regions, we used an in vivo experimental model. Adult male Wistar rats were randomly assigned to one of two conditions, an ad libitum control group or an HFD (60%)-fed group, for eight weeks. At the end of the protocol, glucose and insulin tolerance tests were performed. Additionally, we analyzed the response to a normally innocuous mechanical stimulus and changes in motor coordination. At the end of the protocol, HFD-fed rats presented increased HOMA-IR and metabolic syndrome (MetS) prevalence. HFD-fed rats also developed an increased nociceptive response to mechanical stimuli and neurological injury, resulting in impaired motor function. Hypothalamus and cerebellum neurons from HFD-fed rats presented with nuclear swelling, an absence of nucleoli, and karyolysis. These results reveal that HFD consumption affects vital brain structures such as the cerebellum, hippocampus, and hypothalamus. This, in turn, could be producing neuronal damage, impairing cellular communication, and consequently altering motricity and pain sensitivity. Although direct evidence of a causal link between BBB dysfunction and sensory-motor changes was not observed, understanding the association uncovered in this study could lead to targeted therapeutic strategies.
Collapse
Affiliation(s)
- Laura M. Ubaldo-Reyes
- Department of Anatomy, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Estefania Espitia-Bautista
- Laboratory of Molecular Neurophysiology, National Institute of Psychiatry Ramón de la Fuente, Mexico City 14370, Mexico;
| | - Antonio Barajas-Martínez
- Center for Complexity Science, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Ricardo Martínez-Tapia
- Department of Physiology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.M.-T.); (R.N.-N.); (L.N.)
| | - Verónica Rodríguez-Mata
- Department of Histology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (V.R.-M.); (A.P.-T.)
| | - Roxana Noriega-Navarro
- Department of Physiology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.M.-T.); (R.N.-N.); (L.N.)
| | - Rene Escalona
- Laboratory of Embryology and Genetics, Departamento de Embriología y Genética, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Jesús Castillo-Hernández
- Multidisciplinary Academic Unit Middle Zone, Autonomous University of San Luis Potosí, San Luis Potosí 79615, Mexico;
| | - Armando Pérez-Torres
- Department of Histology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (V.R.-M.); (A.P.-T.)
| | - Luz Navarro
- Department of Physiology, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (R.M.-T.); (R.N.-N.); (L.N.)
| |
Collapse
|
7
|
García-Beltrán A, Lozano Melero A, Martínez Martínez R, Porres Foulquie JM, López Jurado Romero de la Cruz M, Kapravelou G. A Systematic Review of the Beneficial Effects of Berry Extracts on Non-Alcoholic Fatty Liver Disease in Animal Models. Nutr Rev 2024:nuae132. [PMID: 39365946 DOI: 10.1093/nutrit/nuae132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2024] Open
Abstract
CONTEXT Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in Western countries and is strongly associated with several metabolic disorders. Plant-derived bioactive extracts, such as berry extracts, with high antioxidant capacity have been used for the treatment and prevention of this pathology. Moreover, they promote circular economy and sustainability. OBJECTIVE To study the beneficial effects of extracts from different parts of berry plants in animal models of NAFLD. DATA SOURCES A systematic research of the MEDLINE (via PubMed), Cochrane, and Scopus databases was conducted to identify relevant studies published after January 2011. In vivo animal studies of NAFLD were included in which berry extracts of different parts of the plant were administered and significantly improved altered biomarkers related to the pathology, such as lipid metabolism and hepatic steatosis, glucose and glycogen metabolism, and antioxidant and anti-inflammatory biomarkers. DATA EXTRACTION Of a total of 203 articles identified, 31 studies were included after implementation of the inclusion and exclusion criteria. DATA ANALYSIS Most of the studies showed a decrease in steatosis and a stimulation of genes related to β-oxidation and downregulation of lipogenic genes, with administration of berry extracts. Berry extracts also attenuated inflammation and oxidative stress. CONCLUSIONS Administration of berry extracts seems to have promising potential in the design of enriched foodstuffs or nutraceuticals for the treatment of NAFLD.
Collapse
Affiliation(s)
- Alejandro García-Beltrán
- Department of Physiology, Biomedical Research Center, Instituto mixto de Deporte y Salud, University of Granada, 18007 Granada, Spain
| | - Aida Lozano Melero
- Department of Physiology, Biomedical Research Center, Instituto mixto de Deporte y Salud, University of Granada, 18007 Granada, Spain
| | - Rosario Martínez Martínez
- Department of Physiology, Biomedical Research Center, Instituto mixto de Deporte y Salud, University of Granada, 18007 Granada, Spain
| | | | | | - Garyfallia Kapravelou
- Department of Physiology, Faculty of Health Sciences, Campus of Melilla, University of Granada, 52005 Granada, Spain
| |
Collapse
|
8
|
Zhao Y, Qiao M, Wang X, Luo X, Yang J, Hu J. Allantoin reduces glucotoxicity and lipotoxicity in a type 2 diabetes rat model by modulating the PI3K and MAPK signaling pathways. Heliyon 2024; 10:e34716. [PMID: 39144993 PMCID: PMC11320158 DOI: 10.1016/j.heliyon.2024.e34716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
Objective The current study aimed to investigate the potential therapeutic impact of allantoin on diabetes produced by a high-fat diet (HFD) and streptozotocin (STZ) in rats. Subjects and methods Male Sprague-Dawley rats were fed a high-fat diet to induce insulin resistance, followed by streptozotocin injection to induce diabetes. The effect of oral treatment of allantoin (200, 400 and 800 mg/kg/day) for 8 weeks was evaluated by calculating the alteration in metabolic parameters, biochemical indicators, the oral glucose tolerance tests (OGTT) and hyperinsulinemic-euglycemic clamp tests were performed. Histopathological studies were performed in the liver, kidney and pancreas. Next, the expressions of the MAPK and insulin signaling pathway were measured by Western blot analysis to elucidate the potential mechanism underlying these antidiabetic activities. Results The administration of allantoin resulted in a significant decrease in fasting blood glucose (FBG) levels, glycogen levels, and glycosylated hemoglobin levels in diabetic rats. Additionally, allantoin therapy led to a dose-dependent increase in body weight growth and serum insulin levels. In addition, the administration of allantoin resulted in a considerable reduction in lipid profile levels and amelioration of histological alterations in rats with diabetes. The administration of allantoin to diabetic rats resulted in a notable decrease in Malondialdehyde (MDA) levels, accompanied by an increase in the activity of antioxidant enzymes in the serum, liver, and kidney. The findings of oral glucose tolerance and hyperinsulinemic-euglycemic clamp tests demonstrated a significant rise in insulin resistance following the administration of allantoin. The upregulation of IRS-2/PI3K/p-Akt/GLUT expression by allantoin suggests a mechanistic relationship between the PI3K/Akt signaling pathway and the antihyperglycemic activity of allantoin. Furthermore, it resulted in a reduction in the levels of TGF-β1/p38MAPK/Caspase-3 expression in the aforementioned rat tissues affected by diabetes. Conclusions This study implies that allantoin treats type 2 diabetes by activating PI3K. Additionally, it reduces liver, kidney, and pancreatic apoptosis and inflammation-induced insulin resistance.re.
Collapse
Affiliation(s)
- Yao Zhao
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830017, Xinjiang, China
| | - Ming Qiao
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830000, Xinjiang, China
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi 830011, Xinjiang, China
| | - Xiaomei Wang
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830017, Xinjiang, China
| | - Xinjie Luo
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830017, Xinjiang, China
| | - Jianhua Yang
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830000, Xinjiang, China
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi 830011, Xinjiang, China
| | - Junping Hu
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830017, Xinjiang, China
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi 830011, Xinjiang, China
| |
Collapse
|
9
|
Moreira Gobis MDL, Goulart de Souza-Silva T, de Almeida Paula HA. The impact of a western diet on gut microbiota and circadian rhythm: A comprehensive systematic review of in vivo preclinical evidence. Life Sci 2024; 349:122741. [PMID: 38788974 DOI: 10.1016/j.lfs.2024.122741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/13/2024] [Accepted: 05/18/2024] [Indexed: 05/26/2024]
Abstract
AIMS Here, we present a systematic review that compiles in vivo experimental data regarding the effect of the WD on the gut microbiota and its impact on the circadian rhythm. Additionally, we reviewed studies evaluating the combined effects of WD and circadian cycle disruption on gut microbiota and circadian cycle markers. MATERIALS AND METHODS The original studies indexed in PubMed/Medline, Scopus, and Web of Science databases were screened according to the PRISMA strategy. KEY FINDINGS Preclinical studies revealed that WD triggers circadian rhythmicity disruption, reduces the alpha-diversity of the microbiota and favors the growth of bacterial groups that are detrimental to intestinal homeostasis, such as Clostridaceae, Enterococcus, Parasutterella and Proteobacteria. When the WD is combined with circadian clock disruption, gut dysbiosis become more pronounced. Reduced cycling of Per3, Rev-erb and CLOCK in the intestine, which are related to dysregulation of lipid metabolism and potential metabolic disease, was observed. SIGNIFICANCE In conclusion, current evidence supports the potential of WD to trigger microbiota dysregulation, disrupt the biological clock, and increase susceptibility to metabolic disorders and potentially chronic diseases.
Collapse
Affiliation(s)
| | - Thaiany Goulart de Souza-Silva
- Institute of Biological Science, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | |
Collapse
|
10
|
Mennitti LV, de Souza EA, Santamarina AB, Sertorio MN, Jucá A, De Souza DV, Ribeiro DA, Pisani LP. Maternal dietary fatty acid composition and content prior to and during pregnancy and lactation influences serum profile, liver phenotype and hepatic miRNA expression in young male and female offspring. J Nutr Biochem 2024; 129:109639. [PMID: 38583498 DOI: 10.1016/j.jnutbio.2024.109639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/25/2024] [Accepted: 04/03/2024] [Indexed: 04/09/2024]
Abstract
This study aimed to investigate whether modifying the pre-gestational lipid content could mitigate metabolic damage in offspring from dams exposed to a high-fat (HF) diet before conception and during pregnancy and lactation, with a focus on sex-specific outcomes. Specific effects of maternal normolipidic diets on offspring were also assessed. Female Wistar rats received control (C) or HF diets before conception. During pregnancy and lactation, females were distributed in five groups: C-C, HF-HF, HF-C, HF-saturated (HF-S) or HF-polyunsaturated n-3 group (HF-P). Saturated and PUFA n-3 diets were normolipidic. In 21-day-old offspring, corporal parameters, adiposity, serum metabolites, OGTT, liver phenotype, and miR-34a-5p hepatic expression were determined. Pre-gestational HF diet impaired glycemic response in females, independent of any change in body weight. Female and male offspring from dams continuously exposed to HF diet exhibited hyperglycemia, increased adiposity, and disrupted serum lipid profiles. Male offspring showed increased hepatic fat accumulation and miR-34a-5p expression. Shifting maternal dietary lipid content to normolipidic diets restored offspring's phenotype; however, decreased SIRT1, IRβ and IRS1 expression in offspring from dams exposed to HF diet before conception suggested early indicators of glucose metabolism damage. Our findings indicated a pronounced metabolic impact on males. In conclusion, glucose tolerance impairment in females before conception disturbed intrauterine environment, influencing in offspring's phenotype. Modifying maternal dietary lipid content mitigated effects of pre-gestational HF diet exposure on young offspring. Nevertheless, decreased hepatic levels of critical insulin signaling proteins indicated that independently of the maternal diet, pre-existing HF diet-induced glucose intolerance before conception may adversely program the offspring's phenotype.
Collapse
Affiliation(s)
- Laís Vales Mennitti
- Department of Bioscience, Institute of Health and Society, Laboratory of Nutrition and Endocrine Physiology, Federal University of São Paulo, Santos, Brazil; Metabolic Research Laboratories and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| | - Esther Alves de Souza
- Department of Bioscience, Institute of Health and Society, Laboratory of Nutrition and Endocrine Physiology, Federal University of São Paulo, Santos, Brazil
| | - Aline Boveto Santamarina
- Department of Bioscience, Institute of Health and Society, Laboratory of Nutrition and Endocrine Physiology, Federal University of São Paulo, Santos, Brazil
| | - Marcela Nascimento Sertorio
- Department of Bioscience, Institute of Health and Society, Laboratory of Nutrition and Endocrine Physiology, Federal University of São Paulo, Santos, Brazil
| | - Andrea Jucá
- Department of Bioscience, Institute of Health and Society, Laboratory of Nutrition and Endocrine Physiology, Federal University of São Paulo, Santos, Brazil
| | - Daniel Vitor De Souza
- Department of Bioscience, Institute of Health and Society, Laboratory of Nutrition and Endocrine Physiology, Federal University of São Paulo, Santos, Brazil
| | - Daniel Araki Ribeiro
- Department of Bioscience, Institute of Health and Society, Laboratory of Nutrition and Endocrine Physiology, Federal University of São Paulo, Santos, Brazil
| | - Luciana Pellegrini Pisani
- Department of Bioscience, Institute of Health and Society, Laboratory of Nutrition and Endocrine Physiology, Federal University of São Paulo, Santos, Brazil.
| |
Collapse
|
11
|
Doncheva AI, Li Y, Khanal P, Hjorth M, Kolset SO, Norheim FA, Kimmel AR, Dalen KT. Altered hepatic lipid droplet morphology and lipid metabolism in fasted Plin2-null mice. J Lipid Res 2023; 64:100461. [PMID: 37844775 PMCID: PMC10716011 DOI: 10.1016/j.jlr.2023.100461] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/18/2023] Open
Abstract
Perilipin 2 (Plin2) binds to the surface of hepatic lipid droplets (LDs) with expression levels that correlate with triacylglyceride (TAG) content. We investigated if Plin2 is important for hepatic LD storage in fasted or high-fat diet-induced obese Plin2+/+ and Plin2-/- mice. Plin2-/- mice had comparable body weights, metabolic phenotype, glucose tolerance, and circulating TAG and total cholesterol levels compared with Plin2+/+ mice, regardless of the dietary regime. Both fasted and high-fat fed Plin2-/- mice stored reduced levels of hepatic TAG compared with Plin2+/+ mice. Fasted Plin2-/- mice stored fewer but larger hepatic LDs compared with Plin2+/+ mice. Detailed hepatic lipid analysis showed substantial reductions in accumulated TAG species in fasted Plin2-/- mice compared with Plin2+/+ mice, whereas cholesteryl esters and phosphatidylcholines were increased. RNA-Seq revealed minor differences in hepatic gene expression between fed Plin2+/+ and Plin2-/- mice, in contrast to marked differences in gene expression between fasted Plin2+/+ and Plin2-/- mice. Our findings demonstrate that Plin2 is required to regulate hepatic LD size and storage of neutral lipid species in the fasted state, while its role in obesity-induced steatosis is less clear.
Collapse
Affiliation(s)
- Atanaska I Doncheva
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Yuchuan Li
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Hepato-Pancreato-Biliary Surgery, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Prabhat Khanal
- Faculty of Biosciences and Aquaculture, Nord University, Steinkjer, Norway
| | - Marit Hjorth
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Svein O Kolset
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Frode A Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Alan R Kimmel
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, The National Institutes of Health, Bethesda, MD, USA
| | - Knut Tomas Dalen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway; The Norwegian Transgenic Center, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
12
|
Wu HX, Lin X, Cheng CL, Jiang HL, Iqbal J, Liu J, Zhou HD. Fat distribution measurements by chemical shift-encoded transition region extraction predict the risk of hyperglycaemia, dyslipidaemia and metabolic syndrome in mice. NMR IN BIOMEDICINE 2023; 36:e4985. [PMID: 37283179 DOI: 10.1002/nbm.4985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/27/2023] [Accepted: 05/16/2023] [Indexed: 06/08/2023]
Abstract
Metabolically healthy or unhealthy obesity is closely related to metabolic syndrome (MetS). To validate a more accurate diagnostic method for obesity that reflects the risk of metabolic disorders in a pre-clinical mouse model, C57BL/6J mice were fed high-sucrose-high-fat and chow diets for 12 weeks to induce obesity. MRI was performed and analysed by chemical shift-encoded fat-water separation based on the transition region extraction method. Abdominal fat was divided into upper and lower abdominal regions at the horizontal lower border of the liver. Blood samples were collected, and the glucose level, lipid profile, liver function, HbA1c and insulin were tested. k-means clustering and stepwise logistic regression were applied to validate the diagnosis of hyperglycaemia, dyslipidaemia and MetS, and to ascertain the predictive effect of MRI-derived parameters to the metabolic disorders. Pearson or Spearman correlation was used to assess the relationship between MRI-derived parameters and metabolic traits. The receiver-operating characteristic curve was used to evaluate the diagnostic effect of each logistic regression model. A two-sided p value less than 0.05 was considered to indicate statistical significance for all tests. We made the precise diagnosis of obesity, dyslipidaemia, hyperglycaemia and MetS in mice. In all, 14 mice could be diagnosed as having MetS, and the levels of body weight, HbA1c, triglyceride, total cholesterol and low-density lipoprotein cholesterol were significantly higher than in the normal group. Upper abdominal fat better predicted dyslipidaemia (odds ratio, OR = 2.673; area under the receiver-operating characteristic curve, AUCROC = 0.9153) and hyperglycaemia (OR = 2.456; AUCROC = 0.9454), and the abdominal visceral adipose tissue (VAT) was better for predicting MetS risk (OR = 1.187; AUCROC = 0.9619). We identified the predictive effect of fat volume and distribution in dyslipidaemia, hyperglycaemia and MetS. The upper abdominal fat played a better predictive role for the risk of dyslipidaemia and hyperglycaemia, and the abdominal VAT played a better predictive role for the risk of MetS.
Collapse
Affiliation(s)
- Hui-Xuan Wu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiao Lin
- Clinical Research Center for Medical Imaging in Hunan Province, Department of Radiology Quality Control Center in Hunan Province, Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Chuan-Li Cheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Hong-Li Jiang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Junaid Iqbal
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jun Liu
- Clinical Research Center for Medical Imaging in Hunan Province, Department of Radiology Quality Control Center in Hunan Province, Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
13
|
Höpfinger A, Karrasch T, Schäffler A, Schmid A. Circulating Levels of Cathelicidin Antimicrobial Peptide (CAMP) Are Affected by Oral Lipid Ingestion. Nutrients 2023; 15:3021. [PMID: 37447348 DOI: 10.3390/nu15133021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
INTRODUCTION Obesity and related diseases are among the main public health issues in the western world. They are thought to be caused by a state of chronic, low-grade inflammation. Cathelicidin antimicrobial peptide (CAMP) was recently discovered to be expressed and secreted by adipocytes. Representing a novel immunomodulatory adipokine, CAMP might play an important role in the complex interaction between metabolism and inflammation. METHODS In a cohort of 80 volunteers, serum samples were collected prior to, and 2 h, 4 h, and 6 h after, oral lipid ingestion. CAMP, fatty acid binding proteins 2 and 4 (FABP-2/-4), and dipeptidylpeptidase-4 (DPP-4) serum concentrations were measured via ELISA. Human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes were treated with free fatty acids, and gene expression levels of CAMP, FABP-4, and DPP-4 were quantified by RT-PCR. RESULTS The mean base-line CAMP serum concentration was 55.78 ± 29.26 ng/mL, with a range of 10.77-146.24 ng/mL. Interestingly, CAMP serum levels were positively correlated with LDL cholesterol, but negatively correlated with HDL cholesterol and adiponectin. Men exhibited higher CAMP serum concentrations than women, an effect apparently linked to oral contraception in the majority of female participants. In both genders, CAMP serum concentrations significantly decreased in a stepwise manner 4 h and 6 h after oral lipid ingestion. This decline was paralleled by a rise of serum bile acid and triglyceride levels upon lipid ingestion. In human SGBS adipocytes, treatment with free fatty acids did not affect CAMP gene expression, but increased FABP-4 gene expression. CONCLUSIONS In conclusion, systemic levels of the antimicrobial peptide and novel adipokine CAMP are significantly decreased upon oral lipid ingestion. While this decline might be linked to the simultaneous increase in bile acids, the underlying mechanisms remain to be elucidated. Furthermore, CAMP might indicate a putative novel cardiovascular biomarker of both inflammatory and metabolic relevance in metaflammation and adipose inflammation.
Collapse
Affiliation(s)
- Alexandra Höpfinger
- Department of Internal Medicine III, University of Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Thomas Karrasch
- Department of Internal Medicine III, University of Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Andreas Schäffler
- Department of Internal Medicine III, University of Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Andreas Schmid
- Department of Internal Medicine III, University of Giessen, Klinikstr. 33, 35392 Giessen, Germany
| |
Collapse
|
14
|
Mahmood A, Faisal MN, Khan JA, Muzaffar H, Muhammad F, Hussain J, Aslam J, Anwar H. Association of a high-fat diet with I-FABP as a biomarker of intestinal barrier dysfunction driven by metabolic changes in Wistar rats. Lipids Health Dis 2023; 22:68. [PMID: 37237272 DOI: 10.1186/s12944-023-01837-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 05/23/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND The epithelial lining of the gut expresses intestinal fatty-acid binding proteins (I-FABPs), which increase in circulation and in plasma concentration during intestinal damage. From the perspective of obesity, the consumption of a diet rich in fat causes a disruption in the integrity of the gut barrier and an increase in its permeability. HYPOTHESIS There is an association between the expression of I-FABP in the gut and various metabolic changes induced by a high-fat (HF) diet. METHODS Wistar albino rats (n = 90) were divided into three groups (n = 30 per group), viz. One control and two HF diet groups (15 and 30%, respectively) were maintained for 6 weeks. Blood samples were thus collected to evaluate the lipid profile, blood glucose level and other biochemical tests. Tissue sampling was conducted to perform fat staining and immunohistochemistry. RESULTS HF diet-fed rats developed adiposity, insulin resistance, leptin resistance, dyslipidemia, and increased expression of I-FABP in the small intestine compared to the control group. Increased I-FABP expression in the ileal region of the intestine is correlated significantly with higher fat contents in the diet, indicating that higher I-FABP expression occurs due to increased demand of enterocytes to transport lipids, leading to metabolic alterations. CONCLUSION In summary, there is an association between the expression of I-FABP and HF diet-induced metabolic alterations, indicating that I-FABP can be used as a biomarker for intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Aisha Mahmood
- Department of Physiology, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Muhammad Naeem Faisal
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Junaid Ali Khan
- Faculty of Veterinary and Animal Sciences, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Humaira Muzaffar
- Department of Physiology, Government College University, Faisalabad, 38040, Pakistan
| | - Faqir Muhammad
- Faculty of Veterinary Science, Bahaudin Zakariya University, Multan, Pakistan
| | - Jazib Hussain
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jawad Aslam
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Haseeb Anwar
- Department of Physiology, Government College University, Faisalabad, 38040, Pakistan.
| |
Collapse
|
15
|
García-Beltrán A, Martínez R, Porres JM, Arrebola F, Artero IR, Galisteo M, Aranda P, Kapravelou G, López-Jurado M. Novel insights and mechanisms of diet-induced obesity: Mid-term versus long-term effects on hepatic transcriptome and antioxidant capacity in Sprague-Dawley rats. Life Sci 2023; 324:121746. [PMID: 37121540 DOI: 10.1016/j.lfs.2023.121746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/08/2023] [Accepted: 04/25/2023] [Indexed: 05/02/2023]
Abstract
AIMS The study of molecular mechanisms related to obesity and associated pathologies like type 2-diabetes and non-alcoholic fatty liver disease requires animal experimental models in which the type of obesogenic diet and length of the experimental period to induce obesity deeply affect the metabolic alterations. Therefore, this study aimed to test the influence of aging along a rat model of diet-induced obesity in gene expression of the hepatic transcriptome. MAIN METHODS A high-fat/high-fructose diet to induce obesity was used. Mid- (13 weeks) and long-term (21 weeks) periods were established. Caloric intake, bodyweight, hepatic fat, fatty acid profile, histological changes, antioxidant activity, and complete transcriptome were analyzed. KEY FINDINGS Excess bodyweight, hepatic steatosis and altered lipid histology, modifications in liver antioxidant activity, and dysregulated expression of transcripts related to cell structure, glucose & lipid metabolism, antioxidant & detoxifying capacity were found. Modifications in obese and control rats were accounted for by the different lengths of the experimental period studied. SIGNIFICANCE Main mechanisms of hepatic fat accumulation were de novo lipogenesis or altered fatty acid catabolism for mid- or long-term study, respectively. Therefore, the choice of obesity-induction length is a key factor in the model of obesity used as a control for each specific experimental design.
Collapse
Affiliation(s)
- Alejandro García-Beltrán
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Centre for Biomedical Research (CIBM), Institute for Research in Sport and Health (IMUDS), Universidad de Granada, 18016 Granada, Spain
| | - Rosario Martínez
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Centre for Biomedical Research (CIBM), Institute for Research in Sport and Health (IMUDS), Universidad de Granada, 18016 Granada, Spain
| | - Jesus M Porres
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Centre for Biomedical Research (CIBM), Institute for Research in Sport and Health (IMUDS), Universidad de Granada, 18016 Granada, Spain.
| | - Francisco Arrebola
- Department of Histology, Institute of Neurosciences, Centre for Biomedical Research (CIBM), Universidad de Granada, 18016 Granada, Spain
| | - Inmaculada Ruiz Artero
- Department of Histology, Institute of Neurosciences, Centre for Biomedical Research (CIBM), Universidad de Granada, 18016 Granada, Spain
| | - Milagros Galisteo
- Department of Pharmacology, School of Pharmacy, Centre for Biomedical Research (CIBM), Universidad de Granada, Campus Universitario de Cartuja s/n, 18071 Granada, Spain
| | - Pilar Aranda
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Centre for Biomedical Research (CIBM), Institute for Research in Sport and Health (IMUDS), Universidad de Granada, 18016 Granada, Spain
| | - Garyfallia Kapravelou
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Centre for Biomedical Research (CIBM), Institute for Research in Sport and Health (IMUDS), Universidad de Granada, 18016 Granada, Spain
| | - María López-Jurado
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Centre for Biomedical Research (CIBM), Institute for Research in Sport and Health (IMUDS), Universidad de Granada, 18016 Granada, Spain
| |
Collapse
|
16
|
Ağagündüz D, Icer MA, Yesildemir O, Koçak T, Kocyigit E, Capasso R. The roles of dietary lipids and lipidomics in gut-brain axis in type 2 diabetes mellitus. J Transl Med 2023; 21:240. [PMID: 37009872 PMCID: PMC10068184 DOI: 10.1186/s12967-023-04088-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/25/2023] [Indexed: 04/04/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM), one of the main types of Noncommunicable diseases (NCDs), is a systemic inflammatory disease characterized by dysfunctional pancreatic β-cells and/or peripheral insulin resistance, resulting in impaired glucose and lipid metabolism. Genetic, metabolic, multiple lifestyle, and sociodemographic factors are known as related to high T2DM risk. Dietary lipids and lipid metabolism are significant metabolic modulators in T2DM and T2DM-related complications. Besides, accumulated evidence suggests that altered gut microbiota which plays an important role in the metabolic health of the host contributes significantly to T2DM involving impaired or improved glucose and lipid metabolism. At this point, dietary lipids may affect host physiology and health via interaction with the gut microbiota. Besides, increasing evidence in the literature suggests that lipidomics as novel parameters detected with holistic analytical techniques have important roles in the pathogenesis and progression of T2DM, through various mechanisms of action including gut-brain axis modulation. A better understanding of the roles of some nutrients and lipidomics in T2DM through gut microbiota interactions will help develop new strategies for the prevention and treatment of T2DM. However, this issue has not yet been entirely discussed in the literature. The present review provides up-to-date knowledge on the roles of dietary lipids and lipidomics in gut-brain axis in T2DM and some nutritional strategies in T2DM considering lipids- lipidomics and gut microbiota interactions are given.
Collapse
Affiliation(s)
- Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490, Ankara, Turkey.
| | - Mehmet Arif Icer
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Amasya University, 05100, Amasya, Turkey
| | - Ozge Yesildemir
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Bursa Uludag University, 16059, Bursa, Turkey
| | - Tevfik Koçak
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490, Ankara, Turkey
| | - Emine Kocyigit
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ordu University, 52200, Ordu, Turkey
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, Portici, 80055, Naples, Italy.
| |
Collapse
|
17
|
Kietsiriroje N, Shah H, Zare M, O'Mahoney LL, West DJ, Pearson SM, Ajjan RA, Campbell MD. Dietary fat intake is associated with insulin resistance and an adverse vascular profile in patients with T1D: a pooled analysis. Eur J Nutr 2023; 62:1231-1238. [PMID: 36495341 PMCID: PMC10030402 DOI: 10.1007/s00394-022-03070-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Insulin resistance (IR) increases vascular risk in individuals with Type 1 Diabetes (T1D). We aimed to investigate the relationship between dietary intake and IR, as well as vascular biomarkers in T1D. METHODS Baseline data from three randomised controlled trials were pooled. Estimated glucose disposal rate (eGDR) was used as an IR marker. Employing multivariate nutrient density substitution models, we examined the association between macronutrient composition and IR/vascular biomarkers (tumour necrosis factor-α, fibrinogen, tissue factor activity, and plasminogen activator inhibitor-1). RESULTS Of the 107 patients, 50.5% were male with mean age of 29 ± 6 years. Those with lower eGDR were older with a longer diabetes duration, higher insulin requirements, and an adverse vascular profile (p < 0.05). Patients with higher degrees of IR had higher total energy intake (3192 ± 566 vs. 2772 ± 268 vs. 2626 ± 395 kcal/d for eGDR < 5.1 vs. 5.1-8.6 vs. ≥ 8.7 mg/kg/min, p < 0.001) and consumed a higher absolute and proportional amount of fat (47.6 ± 18.6 vs. 30.4 ± 8.1 vs. 25.8 ± 10.4%, p < 0.001). After adjusting for total energy intake, age, sex, and diabetes duration, increased carbohydrate intake offset by an isoenergetic decrease in fat was associated with higher eGDR (β = 0.103, 95% CI 0.044-0.163). In contrast, increased dietary fat at the expense of dietary protein intake was associated with lower eGDR (β = - 0.119, 95% CI - 0.199 to - 0.040). Replacing fat with 5% isoenergetic amount of carbohydrate resulted in decreased vascular biomarkers (p < 0.05). CONCLUSION Higher fat, but not carbohydrate, intake is associated with increased IR and an adverse vascular profile in patients with T1D.
Collapse
Affiliation(s)
- Noppadol Kietsiriroje
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Endocrinology and Metabolism Unit, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Hanya Shah
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, SR1 3SD, UK
| | - Marios Zare
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, SR1 3SD, UK
| | - Lauren L O'Mahoney
- Diabetes Research Centre, Leicester General Hospital, University of Leicester, Leicester, UK
| | - Daniel J West
- Human Nutrition Research Centre, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Sam M Pearson
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Ramzi A Ajjan
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Matthew D Campbell
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK.
- Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland, SR1 3SD, UK.
- Insutiv, T1D One Life Ltd, London, UK.
| |
Collapse
|
18
|
Nota MH, Nicolas S, O’Leary OF, Nolan YM. Outrunning a bad diet: interactions between exercise and a Western-style diet for adolescent mental health, metabolism and microbes. Neurosci Biobehav Rev 2023; 149:105147. [PMID: 36990371 DOI: 10.1016/j.neubiorev.2023.105147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023]
Abstract
Adolescence is a period of biological, psychological and social changes, and the peak time for the emergence of mental health problems. During this life stage, brain plasticity including hippocampal neurogenesis is increased, which is crucial for cognitive functions and regulation of emotional responses. The hippocampus is especially susceptible to environmental and lifestyle influences, mediated by changes in physiological systems, resulting in enhanced brain plasticity but also an elevated risk for developing mental health problems. Indeed, adolescence is accompanied by increased activation of the maturing hypothalamic-pituitary-adrenal axis, sensitivity to metabolic changes due to increased nutritional needs and hormonal changes, and gut microbiota maturation. Importantly, dietary habits and levels of physical activity significantly impact these systems. In this review, the interactions between exercise and Western-style diets, which are high in fat and sugar, on adolescent stress susceptibility, metabolism and the gut microbiota are explored. We provide an overview of current knowledge on implications of these interactions for hippocampal function and adolescent mental health, and speculate on potential mechanisms which require further investigation.
Collapse
|
19
|
Hjorth M, Doncheva A, Norheim F, Ulven SM, Holven KB, Sæther T, Dalen KT. Consumption of salmon fishmeal increases hepatic cholesterol content in obese C57BL/6 J mice. Eur J Nutr 2022; 61:4027-4043. [PMID: 35788891 PMCID: PMC9596588 DOI: 10.1007/s00394-022-02930-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/31/2022] [Indexed: 12/04/2022]
Abstract
PURPOSE By-products from farmed fish contain large amounts of proteins and may be used for human consumption. The purpose of this study was to investigate cardiometabolic effects and metabolic tolerance in mice consuming fishmeal from salmon by-products, salmon filet or beef. METHODS Female C57BL/6J mice were fed chow, as a healthy reference group, or a high-fat diet for 10 weeks to induce obesity and glucose intolerance. Obese mice were subsequently given isocaloric diets containing 50% of the dietary protein from salmon fishmeal, salmon filet or beef for 10 weeks. Mice were subjected to metabolic phenotyping, which included measurements of body composition, energy metabolism in metabolic cages and glucose tolerance. Lipid content and markers of hepatic toxicity were determined in plasma and liver. Hepatic gene and protein expression was determined with RNA sequencing and immunoblotting. RESULTS Mice fed fishmeal, salmon filet or beef had similar food intake, energy consumption, body weight gain, adiposity, glucose tolerance and circulating levels of lipids and hepatic toxicity markers, such as p-ALT and p-AST. Fishmeal increased hepatic cholesterol levels by 35-36% as compared to salmon filet (p = 0.0001) and beef (p = 0.005). This was accompanied by repressed expression of genes involved in steroid and cholesterol metabolism and reduced levels of circulating Pcsk9. CONCLUSION Salmon fishmeal was well tolerated, but increased hepatic cholesterol content. The high cholesterol content in fishmeal may be responsible for the effects on hepatic cholesterol metabolism. Before introducing fishmeal from salmon by-products as a dietary component, it may be advantageous to reduce the cholesterol content in fishmeal.
Collapse
Affiliation(s)
- Marit Hjorth
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Sognsvannsveien 9, Domus Medica, Blindern, P.O. Box 1046, 0317, Oslo, Norway
| | - Atanaska Doncheva
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Sognsvannsveien 9, Domus Medica, Blindern, P.O. Box 1046, 0317, Oslo, Norway
| | - Frode Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Sognsvannsveien 9, Domus Medica, Blindern, P.O. Box 1046, 0317, Oslo, Norway
| | - Stine Marie Ulven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Sognsvannsveien 9, Domus Medica, Blindern, P.O. Box 1046, 0317, Oslo, Norway
| | - Kirsten Bjørklund Holven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Sognsvannsveien 9, Domus Medica, Blindern, P.O. Box 1046, 0317, Oslo, Norway
- Norwegian National Advisory Unit On Familial Hypercholesterolemia, Oslo University Hospital, Aker Sykehus, Postboks 4950, 0424, Oslo, Norway
| | - Thomas Sæther
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372, Oslo, Norway
| | - Knut Tomas Dalen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Sognsvannsveien 9, Domus Medica, Blindern, P.O. Box 1046, 0317, Oslo, Norway.
- The Norwegian Transgenic Center, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
20
|
Kong Y, Niu A, Yuan W, Zhou Y, Xia M, Xiong X, Lu Y, Yin T, Zhang Y, Chen S, Huang Q, Zeng G, Huang Q. Interaction of FOXO1 and SUMOylated PPARγ1 induced by hyperlipidemia and hyperglycemia favors vascular endothelial insulin resistance and dysfunction. Vascul Pharmacol 2022; 147:107125. [PMID: 36252777 DOI: 10.1016/j.vph.2022.107125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/10/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
PPARγ1 and FOXO1 are the key transcription factors that regulate insulin sensitivity. We previously found that a small ubiquitin-related modifier of PPARγ1 at K77 (SUMOylation) favored endothelial insulin resistance (IR) induced by high fat/high glucose (HF/HG) administration. However, whether and how the crosstalk between SUMOylated PPARγ1 and FOXO1 would mediate the development of the endothelial IR and dysfunction remains unclear. Here, we emphasize how PPARγ1-K77 SUMOylation would interact with FOXO1 and participate in the development of the endothelial IR and dysfunction. Our results show that the combination of HF/HG and PPARγ1-K77 SUMOylation exhibits a synergistic deteriorative effect on the endothelial IR and dysfunction, presenting decreased NO levels and elevated ET-1 levels, weakened PI3K/Akt/eNOS signaling, and impaired endothelium-dependent vasodilation function. The further researches reveal that PPARγ1-K77 SUMOylation readily interacts with FOXO1, and FOXO1 occupies the PPAR response element (PPRE) which is supposed to be occupied by PPARγ, thus resulting in the decrease of PPARγ1 transcription activity and the mitigation of the PI3K/Akt signaling. Moreover, the mitigation of the PI3K/Akt signaling promotes in turn the accumulation of FOXO1 in the nucleus where FOXO1 interacts with the SUMOylated PPARγ1, thus exerting a positive feedback effect on IR pathogenesis. The findings uncover a novel association between PPARγ1-K77 SUMOylation and FOXO1, which contributes to our understanding of the pathogenesis of endothelial IR and dysfunction and provides novel pharmacological targets for diabetic angiopathy.
Collapse
Affiliation(s)
- Ying Kong
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China; Department of Pharmacy, the First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Ailin Niu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Wanwan Yuan
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Yumeng Zhou
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Min Xia
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Xiaowei Xiong
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Yanli Lu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Tingting Yin
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Yanan Zhang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Sheng Chen
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Qianqian Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Guohua Zeng
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Qiren Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, 330006, Jiangxi, PR China.
| |
Collapse
|
21
|
Restricted cafeteria feeding and treadmill exercise improved body composition, metabolic profile and exploratory behavior in obese male rats. Sci Rep 2022; 12:19545. [PMID: 36379981 PMCID: PMC9666649 DOI: 10.1038/s41598-022-23464-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to evaluate, in male Long-Evans rats, whether a restricted-cafeteria diet (CAFR), based on a 30% calorie restriction vs continuous ad libitum cafeteria (CAF) fed animals, administered alone or in combination with moderate treadmill exercise (12 m/min, 35 min, 5 days/week for 8 weeks), was able to ameliorate obesity and the associated risk factors induced by CAF feeding for 18 weeks and to examine the changes in circadian locomotor activity, hypothalamic-pituitary-adrenal (HPA) axis functionality, and stress response elicited by this dietary pattern. In addition to the expected increase in body weight and adiposity, and the development of metabolic dysregulations compatible with Metabolic Syndrome, CAF intake resulted in a sedentary profile assessed by the home-cage activity test, reduced baseline HPA axis activity through decreased corticosterone levels, and boosted exploratory behavior. Both CAFR alone and in combination with exercise reduced abdominal adiposity and hypercholesterolemia compared to CAF. Exercise increased baseline locomotor activity in the home-cage in all dietary groups, boosted exploratory behavior in STD and CAF, partially decreased anxiety-like behavior in CAF and CAFR, but did not affect HPA axis-related parameters.
Collapse
|
22
|
Cummings J, Ortiz A, Castellino J, Kinney J. Diabetes: Risk factor and translational therapeutic implications for Alzheimer's disease. Eur J Neurosci 2022; 56:5727-5757. [PMID: 35128745 PMCID: PMC9393901 DOI: 10.1111/ejn.15619] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 12/31/2022]
Abstract
Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) commonly co-occur. T2DM increases the risk for AD by approximately twofold. Animal models provide one means of interrogating the relationship of T2DM to AD and investigating brain insulin resistance in the pathophysiology of AD. Animal models show that persistent hyperglycaemia results in chronic low-grade inflammation that may contribute to the development of neuroinflammation and accelerate the pathobiology of AD. Epidemiological studies suggest that patients with T2DM who received treatment with specific anti-diabetic agents have a decreased risk for the occurrence of AD and all-cause dementia. Agents such as metformin ameliorate T2DM and may have other important systemic effects that lower the risk of AD. Glucagon-like peptide 1 (GLP-1) agonists have been associated with a decreased risk for AD in patients with T2DM. Both insulin and non-insulin anti-diabetic treatments have been evaluated for the treatment of AD in clinical trials. In most cases, patients included in the trials have clinical features of AD but do not have T2DM. Many of the trials were conducted prior to the use of diagnostic biomarkers for AD. Trials have had a wide range of durations and population sizes. Many of the agents used to treat T2DM do not cross the blood brain barrier, and the effects are posited to occur via lowering of peripheral hyperglycaemia and reduction of peripheral and central inflammation. Clinical trials of anti-diabetic agents to treat AD are ongoing and will provide insight into the therapeutic utility of these agents.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Chambers‐Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | - Andrew Ortiz
- Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | | | - Jefferson Kinney
- Chambers‐Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA,Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| |
Collapse
|
23
|
Meneses MJ, Sousa-Lima I, Jarak I, Raposo JF, Alves MG, Macedo MP. Distinct impacts of fat and fructose on the liver, muscle, and adipose tissue metabolome: An integrated view. Front Endocrinol (Lausanne) 2022; 13:898471. [PMID: 36060961 PMCID: PMC9428722 DOI: 10.3389/fendo.2022.898471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Objective In the last years, changes in dietary habits have contributed to the increasing prevalence of metabolic disorders, such as non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM). The differential burden of lipids and fructose on distinct organs needs to be unveiled. Herein, we hypothesized that high-fat and high-fructose diets differentially affect the metabolome of insulin-sensitive organs such as the liver, muscle, and different adipose tissue depots. Methods We have studied the impact of 12 weeks of a control (11.50% calories from fat, 26.93% from protein, and 61.57% from carbohydrates), high-fat/sucrose (HFat), or high-fructose (HFruct) feeding on C57Bl/6J male mice. Besides glucose homeostasis, we analyzed the hepatic levels of glucose and lipid-metabolism-related genes and the metabolome of the liver, the muscle, and white (WAT) and brown adipose tissue (BAT) depots. Results HFat diet led to a more profound impact on hepatic glucose and lipid metabolism than HFruct, with mice presenting glucose intolerance, increased saturated fatty acids, and no glycogen pool, yet both HFat and HFruct presented hepatic insulin resistance. HFat diet promoted a decrease in glucose and lactate pools in the muscle and an increase in glutamate levels. While HFat had alterations in BAT metabolites that indicate increased thermogenesis, HFruct led to an increase in betaine, a protective metabolite against fructose-induced inflammation. Conclusions Our data illustrate that HFat and HFruct have a negative but distinct impact on the metabolome of the liver, muscle, WAT, and BAT.
Collapse
Affiliation(s)
- Maria João Meneses
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
- Portuguese Diabetes Association - Education and Research Center (APDP-ERC), Lisbon, Portugal
| | - Inês Sousa-Lima
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
| | - Ivana Jarak
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - João F. Raposo
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
- Portuguese Diabetes Association - Education and Research Center (APDP-ERC), Lisbon, Portugal
| | - Marco G. Alves
- Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Maria Paula Macedo
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
- Portuguese Diabetes Association - Education and Research Center (APDP-ERC), Lisbon, Portugal
- Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
24
|
Li Z, Gurung M, Rodrigues RR, Padiadpu J, Newman NK, Manes NP, Pederson JW, Greer RL, Vasquez-Perez S, You H, Hioki KA, Moulton Z, Fel A, De Nardo D, Dzutsev AK, Nita-Lazar A, Trinchieri G, Shulzhenko N, Morgun A. Microbiota and adipocyte mitochondrial damage in type 2 diabetes are linked by Mmp12+ macrophages. J Exp Med 2022; 219:213260. [PMID: 35657352 PMCID: PMC9170383 DOI: 10.1084/jem.20220017] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/22/2022] [Accepted: 05/05/2022] [Indexed: 01/07/2023] Open
Abstract
Microbiota contribute to the induction of type 2 diabetes by high-fat/high-sugar (HFHS) diet, but which organs/pathways are impacted by microbiota remain unknown. Using multiorgan network and transkingdom analyses, we found that microbiota-dependent impairment of OXPHOS/mitochondria in white adipose tissue (WAT) plays a primary role in regulating systemic glucose metabolism. The follow-up analysis established that Mmp12+ macrophages link microbiota-dependent inflammation and OXPHOS damage in WAT. Moreover, the molecular signature of Mmp12+ macrophages in WAT was associated with insulin resistance in obese patients. Next, we tested the functional effects of MMP12 and found that Mmp12 genetic deficiency or MMP12 inhibition improved glucose metabolism in conventional, but not in germ-free mice. MMP12 treatment induced insulin resistance in adipocytes. TLR2-ligands present in Oscillibacter valericigenes bacteria, which are expanded by HFHS, induce Mmp12 in WAT macrophages in a MYD88-ATF3-dependent manner. Thus, HFHS induces Mmp12+ macrophages and MMP12, representing a microbiota-dependent bridge between inflammation and mitochondrial damage in WAT and causing insulin resistance.
Collapse
Affiliation(s)
- Zhipeng Li
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR,Shanghai Mengniu Biotechnology R&D Co., Ltd., Shanghai, China
| | - Manoj Gurung
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR
| | - Richard R. Rodrigues
- College of Pharmacy, Oregon State University, Corvallis, OR,Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD,Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD
| | | | | | - Nathan P. Manes
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Jacob W. Pederson
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR
| | - Renee L. Greer
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR
| | | | - Hyekyoung You
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR
| | - Kaito A. Hioki
- College of Pharmacy, Oregon State University, Corvallis, OR
| | - Zoe Moulton
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR
| | - Anna Fel
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Dominic De Nardo
- Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Amiran K. Dzutsev
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Aleksandra Nita-Lazar
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Giorgio Trinchieri
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD,Giorgio Trinchieri:
| | - Natalia Shulzhenko
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR,Correspondence to Natalia Shulzhenko:
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, Corvallis, OR,Andrey Morgun:
| |
Collapse
|
25
|
Boonloh K, Thanaruksa R, Proongkhong T, Thawornchinsombut S, Pannangpetch P. Nil-Surin Rice Bran Hydrolysates Improve Lipid Metabolism and Hepatic Steatosis by Regulating Secretion of Adipokines and Expression of Lipid-Metabolism Genes. J Med Food 2022; 25:597-606. [PMID: 35708630 DOI: 10.1089/jmf.2021.k.0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Overconsumption of a high caloric diet is associated with metabolic disorders and a heightened risk of diabetes mellitus (DM), hepatic steatosis, and cardiovascular complications. The use of functional food has received much attention as a strategy in the prevention and treatment of metabolic disorders. This present study investigated whether Nil-Surin rice bran hydrolysates (NRH) could prevent or ameliorate the progression of metabolic disorders in rats in which insulin resistance (IR) was induced by a high fat-high fructose diet (HFFD). After 10 weeks of the HFFD, the rats showed elevated fasting blood glucose (FBG), impaired glucose tolerance, dysregulation of adipokine secretion, distorted lipid metabolism such as dyslipidemia, and increased intrahepatic fat accumulation. The IR was significantly attenuated by a daily dose of NRH (100 or 300 mg/kg/day). Doses of NRH rectified adipokine dysregulation by increasing serum adiponectin and improving hyperleptinemia. Interestingly, NRH decreased intrahepatic fat accumulation and improved dyslipidemia as shown by decreased levels of hepatic triglyceride (TG) and serum TG, total cholesterol and low-density lipoprotein cholesterol, and increased high-density lipoprotein cholesterol. In addition, a modulation of expression of lipid metabolism genes was observed: NRH prevented upregulation of the lipogenesis genes Srebf1 and Fasn. In addition, NRH enhanced the expression of fatty-acid oxidation genes, as evidenced by an increase of Ppara and Cpt1a when compared with the HFFD control group. The activities of NRH in the modulation of lipid metabolism and rectifying the dysregulation of adipokines may result in a decreased risk of DM and hepatic steatosis. Therefore, NRH may be beneficial in ameliorating metabolic disorders in the HFFD model.
Collapse
Affiliation(s)
- Kampeebhorn Boonloh
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Muang District, Thailand.,Cardiovascular Research Group, Khon Kaen University, Muang District, Thailand
| | - Ratthipha Thanaruksa
- Rice Department, Surin Rice Research Center, Agricultural and Cooperatives Ministry, Surin, Thailand
| | - Tunvaraporn Proongkhong
- Rice Department, Chum Phae Rice Research Center, Agricultural and Cooperatives Ministry, Khon Kaen, Thailand
| | - Supawan Thawornchinsombut
- Department of Food Technology, Faculty of Technology, Khon Kaen University, Muang District, Thailand
| | - Patchareewan Pannangpetch
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Muang District, Thailand.,Cardiovascular Research Group, Khon Kaen University, Muang District, Thailand
| |
Collapse
|
26
|
Rodrigo N, Saad S, Pollock C, Glastras SJ. Diet Modification before or during Pregnancy on Maternal and Foetal Outcomes in Rodent Models of Maternal Obesity. Nutrients 2022; 14:2154. [PMID: 35631295 PMCID: PMC9146671 DOI: 10.3390/nu14102154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 12/10/2022] Open
Abstract
The obesity epidemic has serious implications for women of reproductive age; its rising incidence is associated not just with health implications for the mother but also has transgenerational ramifications for the offspring. Increased incidence of diabetes, cardiovascular disease, obesity, and kidney disease are seen in both the mothers and the offspring. Animal models, such as rodent studies, are fundamental to studying maternal obesity and its impact on maternal and offspring health, as human studies lack rigorous controlled experimental design. Furthermore, the short and prolific reproductive potential of rodents enables examination across multiple generations and facilitates the exploration of interventional strategies to mitigate the impact of maternal obesity, both before and during pregnancy. Given that obesity is a major public health concern, it is important to obtain a greater understanding of its pathophysiology and interaction with reproductive health, placental physiology, and foetal development. This narrative review focuses on the known effects of maternal obesity on the mother and the offspring, and the benefits of interventional strategies, including dietary intervention, before or during pregnancy on maternal and foetal outcomes. It further examines the contribution of rodent models of maternal obesity to elucidating pathophysiological pathways of disease development, as well as methods to reduce the impact of obesity on the mothers and the developing foetus. The translation of these findings into the human experience will also be discussed.
Collapse
Affiliation(s)
- Natassia Rodrigo
- Department of Diabetes, Endocrinology and Metabolism, Royal North Shore Hospital, Sydney 2065, Australia;
- Kolling Institute of Medical Research, Sydney 2065, Australia; (S.S.); (C.P.)
- Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
| | - Sonia Saad
- Kolling Institute of Medical Research, Sydney 2065, Australia; (S.S.); (C.P.)
- Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
| | - Carol Pollock
- Kolling Institute of Medical Research, Sydney 2065, Australia; (S.S.); (C.P.)
- Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
- Department of Renal Medicine, Royal North Shore Hospital, Sydney 2065, Australia
| | - Sarah J. Glastras
- Department of Diabetes, Endocrinology and Metabolism, Royal North Shore Hospital, Sydney 2065, Australia;
- Kolling Institute of Medical Research, Sydney 2065, Australia; (S.S.); (C.P.)
- Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
| |
Collapse
|
27
|
Zakharova IO, Bayunova LV, Derkach KV, Ilyasov IO, Shpakov AO, Avrova NF. Effects of Intranasally Administered Insulin and Gangliosides on Metabolic Parameters and Activity of the Hepatic Insulin System in Rats with Type 2 Diabetes Mellitus. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022020077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
28
|
Mietelska-Porowska A, Domańska J, Want A, Więckowska-Gacek A, Chutorański D, Koperski M, Wojda U. Induction of Brain Insulin Resistance and Alzheimer's Molecular Changes by Western Diet. Int J Mol Sci 2022; 23:ijms23094744. [PMID: 35563135 PMCID: PMC9102094 DOI: 10.3390/ijms23094744] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
The term Western diet (WD) describes the consumption of large amounts of highly processed foods, rich in simple sugars and saturated fats. Long-term WD feeding leads to insulin resistance, postulated as a risk factor for Alzheimer’s disease (AD). AD is the main cause of progressive dementia characterized by the deposition of amyloid-β (Aβ) plaques and neurofibrillary tangles consisting of the hyperphosphorylated tau (p-Tau) protein in the brain, starting from the entorhinal cortex and the hippocampus. In this study, we report that WD-derived impairment in insulin signaling induces tau and Aβ brain pathology in wild-type C57BL/6 mice, and that the entorhinal cortex is more sensitive than the hippocampus to the impairment of brain insulin signaling. In the brain areas developing WD-induced insulin resistance, we observed changes in p-Tau(Thr231) localization in neuronal subcellular compartments, indicating progressive tauopathy, and a decrease in amyloid precursor protein levels correlating with the appearance of Aβ peptides. These results suggest that WD promotes the development of AD and may be considered not only a risk factor, but also a modifiable trigger of AD.
Collapse
|
29
|
Pellegrinelli V, Rodriguez-Cuenca S, Rouault C, Figueroa-Juarez E, Schilbert H, Virtue S, Moreno-Navarrete JM, Bidault G, Vázquez-Borrego MC, Dias AR, Pucker B, Dale M, Campbell M, Carobbio S, Lin YH, Vacca M, Aron-Wisnewsky J, Mora S, Masiero MM, Emmanouilidou A, Mukhopadhyay S, Dougan G, den Hoed M, Loos RJF, Fernández-Real JM, Chiarugi D, Clément K, Vidal-Puig A. Dysregulation of macrophage PEPD in obesity determines adipose tissue fibro-inflammation and insulin resistance. Nat Metab 2022; 4:476-494. [PMID: 35478031 PMCID: PMC7617220 DOI: 10.1038/s42255-022-00561-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 03/18/2022] [Indexed: 02/02/2023]
Abstract
Resulting from impaired collagen turnover, fibrosis is a hallmark of adipose tissue (AT) dysfunction and obesity-associated insulin resistance (IR). Prolidase, also known as peptidase D (PEPD), plays a vital role in collagen turnover by degrading proline-containing dipeptides but its specific functional relevance in AT is unknown. Here we show that in human and mouse obesity, PEPD expression and activity decrease in AT, and PEPD is released into the systemic circulation, which promotes fibrosis and AT IR. Loss of the enzymatic function of PEPD by genetic ablation or pharmacological inhibition causes AT fibrosis in mice. In addition to its intracellular enzymatic role, secreted extracellular PEPD protein enhances macrophage and adipocyte fibro-inflammatory responses via EGFR signalling, thereby promoting AT fibrosis and IR. We further show that decreased prolidase activity is coupled with increased systemic levels of PEPD that act as a pathogenic trigger of AT fibrosis and IR. Thus, PEPD produced by macrophages might serve as a biomarker of AT fibro-inflammation and could represent a therapeutic target for AT fibrosis and obesity-associated IR and type 2 diabetes.
Collapse
Affiliation(s)
- V Pellegrinelli
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK.
| | - S Rodriguez-Cuenca
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
- Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, P. R. China
| | - C Rouault
- Sorbonne University, INSERM, NutriOmique Research Unit, Paris, France
| | - E Figueroa-Juarez
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - H Schilbert
- Genetics and Genomics of Plants, Centre for Biotechnology (CeBiTec) & Faculty of Biology, Bielefeld University, Bielefeld, Germany
| | - S Virtue
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - J M Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Girona Biomedical Research Institute (IDIBGI), University Hospital of Girona Dr Josep Trueta, Girona, Spain
- Department of Medicine, University of Girona, Girona, Spain
- CIBERobn Pathophysiology of Obesity and Nutrition, Institut of Salud Carlos III, Madrid, Spain
| | - G Bidault
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - M C Vázquez-Borrego
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain
| | - A R Dias
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - B Pucker
- Genetics and Genomics of Plants, Centre for Biotechnology (CeBiTec) & Faculty of Biology, Bielefeld University, Bielefeld, Germany
- Evolution and Diversity, Department of Plant Sciences, University of Cambridge, Cambridge, UK
| | - M Dale
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - M Campbell
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
- Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, P. R. China
| | - S Carobbio
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
- Centro de Investigacion Principe Felipe, Valencia, Spain
| | - Y H Lin
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
- Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - M Vacca
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
- Insterdisciplinary Department of Medicine, Università degli Studi di Bari 'Aldo Moro', Bari, Italy
| | - J Aron-Wisnewsky
- Sorbonne University, INSERM, NutriOmique Research Unit, Paris, France
- Assistance-Publique Hôpitaux de Paris, Nutrition department, Pitié-Salpêtrière hospital, Paris, France
| | - S Mora
- Dept Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine, University of Barcelona (IBUB), Barcelona, Spain
| | - M M Masiero
- The Beijer Laboratory and Department of Immunology, Genetics and Pathology, Uppsala University and SciLifeLab, Uppsala, Sweden
| | - A Emmanouilidou
- The Beijer Laboratory and Department of Immunology, Genetics and Pathology, Uppsala University and SciLifeLab, Uppsala, Sweden
| | - S Mukhopadhyay
- MRC Centre for Transplantation Peter Gorer Department of Immunobiology School of Immunology & Microbial Sciences King's College, London, UK
| | - G Dougan
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Division of Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge, UK
| | - M den Hoed
- The Beijer Laboratory and Department of Immunology, Genetics and Pathology, Uppsala University and SciLifeLab, Uppsala, Sweden
| | - R J F Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - J M Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Girona Biomedical Research Institute (IDIBGI), University Hospital of Girona Dr Josep Trueta, Girona, Spain
- Department of Medicine, University of Girona, Girona, Spain
- CIBERobn Pathophysiology of Obesity and Nutrition, Institut of Salud Carlos III, Madrid, Spain
| | - D Chiarugi
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - K Clément
- Sorbonne University, INSERM, NutriOmique Research Unit, Paris, France
- Assistance-Publique Hôpitaux de Paris, Nutrition department, Pitié-Salpêtrière hospital, Paris, France
| | - A Vidal-Puig
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, UK.
- Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, P. R. China.
- Centro de Investigacion Principe Felipe, Valencia, Spain.
| |
Collapse
|
30
|
Yokota‐Nakagi N, Omoto S, Tazumi S, Kawakami M, Takamata A, Morimoto K. Estradiol replacement improves high-fat diet-induced insulin resistance in ovariectomized rats. Physiol Rep 2022; 10:e15193. [PMID: 35238495 PMCID: PMC8892597 DOI: 10.14814/phy2.15193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 05/15/2023] Open
Abstract
The role of 17β-estradiol (E2) in high-fat diet (HFD)-induced alteration of the protein kinase B (Akt) signaling pathway in ovariectomized (OVX) rats is unclear. Therefore, we examined whether chronic estrogen replacement restores HFD-induced impairment in insulin sensitivity by its effects concomitant with alterations in the Akt isoform 2 (Akt2) and Akt substrate of 160 kDa (AS160) phosphorylation in muscles of OVX rats. Nine-week-old female Wistar rats underwent ovariectomy under anesthesia; after 4 weeks, subcutaneous implantation of either E2 or placebo (PL) pellets was performed, and HFD feeding was initiated. Intravenous glucose tolerance tests were performed to assess insulin sensitivity. Following insulin injection into rats' portal vein, the liver and gastrocnemius muscle were dissected for insulin signaling analysis. We observed that HFD increased energy intake and body weight in the PL group; however, it was temporarily decreased in the E2 group. Adipose tissue accumulation was larger in HFD-fed rats than in normal chow diet (NCD)-fed rats in the PL group; however, this difference was not observed in the E2 group. HFD reduced insulin sensitivity in the PL group only. In vivo insulin stimulation increased Akt2 phosphorylation in the muscles of NCD-fed rats in both groups. In contrast, HFD affected insulin-stimulated phosphorylation of Akt2 and AS160 in the muscles of rats in the PL group but not in the E2 group. Our data suggest that E2 replacement improves HFD-induced insulin resistance, and this effect is accompanied by the alterations in the Akt2 and AS160 phosphorylation in insulin-stimulated muscles of OVX rats.
Collapse
Affiliation(s)
- Naoko Yokota‐Nakagi
- Department of Environmental HealthFaculty of Human Life and EnvironmentNara Women’s UniversityNaraJapan
- Department of Health and NutritionFaculty of Health ScienceKyoto Koka Women’s UniversityKyotoJapan
| | - Sayo Omoto
- Department of Environmental HealthFaculty of Human Life and EnvironmentNara Women’s UniversityNaraJapan
| | - Shoko Tazumi
- Department of Environmental HealthFaculty of Human Life and EnvironmentNara Women’s UniversityNaraJapan
| | - Mizuho Kawakami
- Department of Environmental HealthFaculty of Human Life and EnvironmentNara Women’s UniversityNaraJapan
| | - Akira Takamata
- Department of Environmental HealthFaculty of Human Life and EnvironmentNara Women’s UniversityNaraJapan
| | - Keiko Morimoto
- Department of Environmental HealthFaculty of Human Life and EnvironmentNara Women’s UniversityNaraJapan
- Department of Health and NutritionFaculty of Health ScienceKyoto Koka Women’s UniversityKyotoJapan
| |
Collapse
|
31
|
Parker J, O’Brien C, Hawrelak J, Gersh FL. Polycystic Ovary Syndrome: An Evolutionary Adaptation to Lifestyle and the Environment. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19031336. [PMID: 35162359 PMCID: PMC8835454 DOI: 10.3390/ijerph19031336] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023]
Abstract
Polycystic ovary syndrome (PCOS) is increasingly recognized as a complex metabolic disorder that manifests in genetically susceptible women following a range of negative exposures to nutritional and environmental factors related to contemporary lifestyle. The hypothesis that PCOS phenotypes are derived from a mismatch between ancient genetic survival mechanisms and modern lifestyle practices is supported by a diversity of research findings. The proposed evolutionary model of the pathogenesis of PCOS incorporates evidence related to evolutionary theory, genetic studies, in utero developmental epigenetic programming, transgenerational inheritance, metabolic features including insulin resistance, obesity and the apparent paradox of lean phenotypes, reproductive effects and subfertility, the impact of the microbiome and dysbiosis, endocrine-disrupting chemical exposure, and the influence of lifestyle factors such as poor-quality diet and physical inactivity. Based on these premises, the diverse lines of research are synthesized into a composite evolutionary model of the pathogenesis of PCOS. It is hoped that this model will assist clinicians and patients to understand the importance of lifestyle interventions in the prevention and management of PCOS and provide a conceptual framework for future research. It is appreciated that this theory represents a synthesis of the current evidence and that it is expected to evolve and change over time.
Collapse
Affiliation(s)
- Jim Parker
- School of Medicine, University of Wollongong, Wollongong 2500, Australia
- Correspondence:
| | - Claire O’Brien
- Faculty of Science and Technology, University of Canberra, Bruce 2617, Australia;
| | - Jason Hawrelak
- College of Health and Medicine, University of Tasmania, Hobart 7005, Australia;
| | - Felice L. Gersh
- College of Medicine, University of Arizona, Tucson, AZ 85004, USA;
| |
Collapse
|
32
|
Merz KE, Hwang J, Zhou C, Veluthakal R, McCown EM, Hamilton A, Oh E, Dai W, Fueger PT, Jiang L, Huss JM, Thurmond DC. Enrichment of the exocytosis protein STX4 in skeletal muscle remediates peripheral insulin resistance and alters mitochondrial dynamics via Drp1. Nat Commun 2022; 13:424. [PMID: 35058456 PMCID: PMC8776765 DOI: 10.1038/s41467-022-28061-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 01/05/2022] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial dysfunction is implicated in skeletal muscle insulin resistance. Syntaxin 4 (STX4) levels are reduced in human diabetic skeletal muscle, and global transgenic enrichment of STX4 expression improves insulin sensitivity in mice. Here, we show that transgenic skeletal muscle-specific STX4 enrichment (skmSTX4tg) in mice reverses established insulin resistance and improves mitochondrial function in the context of diabetogenic stress. Specifically, skmSTX4tg reversed insulin resistance caused by high-fat diet (HFD) without altering body weight or food consumption. Electron microscopy of wild-type mouse muscle revealed STX4 localisation at or proximal to the mitochondrial membrane. STX4 enrichment prevented HFD-induced mitochondrial fragmentation and dysfunction through a mechanism involving STX4-Drp1 interaction and elevated AMPK-mediated phosphorylation at Drp1 S637, which favors fusion. Our findings challenge the dogma that STX4 acts solely at the plasma membrane, revealing that STX4 localises at/proximal to and regulates the function of mitochondria in muscle. These results establish skeletal muscle STX4 enrichment as a candidate therapeutic strategy to reverse peripheral insulin resistance.
Collapse
Affiliation(s)
- Karla E Merz
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA, USA
- Amgen, Thousand Oaks, CA, USA
| | - Jinhee Hwang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Chunxue Zhou
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Rajakrishnan Veluthakal
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Erika M McCown
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Angelica Hamilton
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Eunjin Oh
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Wenting Dai
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Patrick T Fueger
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
- Comprehensive Metabolic Phenotyping Core, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Lei Jiang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Janice M Huss
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
- Washington University School of Medicine, St. Louis, MO, USA
| | - Debbie C Thurmond
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
33
|
Grzęda E, Matuszewska J, Ziarniak K, Gertig-Kolasa A, Krzyśko- Pieczka I, Skowrońska B, Sliwowska JH. Animal Foetal Models of Obesity and Diabetes - From Laboratory to Clinical Settings. Front Endocrinol (Lausanne) 2022; 13:785674. [PMID: 35197931 PMCID: PMC8858803 DOI: 10.3389/fendo.2022.785674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/11/2022] [Indexed: 12/26/2022] Open
Abstract
The prenatal period, during which a fully formed newborn capable of surviving outside its mother's body is built from a single cell, is critical for human development. It is also the time when the foetus is particularly vulnerable to environmental factors, which may modulate the course of its development. Both epidemiological and animal studies have shown that foetal programming of physiological systems may alter the growth and function of organs and lead to pathology in adulthood. Nutrition is a particularly important environmental factor for the pregnant mother as it affects the condition of offspring. Numerous studies have shown that an unbalanced maternal metabolic status (under- or overnutrition) may cause long-lasting physiological and behavioural alterations, resulting in metabolic disorders, such as obesity and type 2 diabetes (T2DM). Various diets are used in laboratory settings in order to induce maternal obesity and metabolic disorders, and to alter the offspring development. The most popular models are: high-fat, high-sugar, high-fat-high-sugar, and cafeteria diets. Maternal undernutrition models are also used, which results in metabolic problems in offspring. Similarly to animal data, human studies have shown the influence of mothers' diets on the development of children. There is a strong link between the maternal diet and the birth weight, metabolic state, changes in the cardiovascular and central nervous system of the offspring. The mechanisms linking impaired foetal development and adult diseases remain under discussion. Epigenetic mechanisms are believed to play a major role in prenatal programming. Additionally, sexually dimorphic effects on offspring are observed. Therefore, further research on both sexes is necessary.
Collapse
Affiliation(s)
- Emilia Grzęda
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
| | - Julia Matuszewska
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
| | - Kamil Ziarniak
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
- Molecular and Cell Biology Unit, Poznań University of Medical Sciences, Poznań, Poland
| | - Anna Gertig-Kolasa
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Izabela Krzyśko- Pieczka
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Bogda Skowrońska
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Joanna H. Sliwowska
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
- *Correspondence: Joanna H. Sliwowska,
| |
Collapse
|
34
|
Yoshimura A, Yamaguchi T, Kugita M, Kumamoto K, Shiogama K, Ogitsu N, Yoneda M, Miura T, Nagamura Y, Nagao S. High Levels of Dietary Lard or Sucrose May Aggravate Lysosomal Renal Injury in Non-Obese, Streptozotocin-Injected CD-1 Mice Provided Isocaloric Diets. J Nutr Sci Vitaminol (Tokyo) 2021; 67:243-248. [PMID: 34470999 DOI: 10.3177/jnsv.67.243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Daily fat and sugar intake has increased in Japan, while total energy intake has decreased. However, the number of type 2 diabetes mellitus patients has increased, and this often causes renal injury characterized by autophagic vacuoles. Although many studies with comparisons of high fat or sugar versus a normal macronutrient balanced diet have been reported, there are few studies that equalized calorie intake and body weights. In the current study, AIN93M diets (CONT group) with matching energy content with lard derived high saturated fat (LARD group), soybean oil derived unsaturated fat (SOY OIL group) and sucrose (SUCROSE group) were provided to compare their effects on renal morphology in streptozotocin-injected CD-1 mice without causing obesity. The number of renal tubular vacuoles was higher in SUCROSE and slightly higher in LARD compared with CONT mice, and was higher in LARD and SUCROSE compared with SOY OIL mice. Most of those vacuoles were LAMP1-positive, a marker of lysosomal autophagy. These results suggest that despite identical energy contents, diets with high sucrose or saturated fat compared to unsaturated fat may aggravate lysosomal renal injury in a non-obese, streptozotocin-induced model of diabetes mellitus.
Collapse
Affiliation(s)
- Aya Yoshimura
- Education and Research Center of Animal Models for Human Diseases, Fujita Health University
| | - Tamio Yamaguchi
- Department of Clinical Nutrition, Faculty of Health Science, Suzuka University of Medical Science
| | - Masanori Kugita
- Education and Research Center of Animal Models for Human Diseases, Fujita Health University
| | - Kanako Kumamoto
- Education and Research Center of Animal Models for Human Diseases, Fujita Health University
| | - Kazuya Shiogama
- Division of Morphology and Cell Function, Faculty of Medical Technology, School of Health Sciences, Fujita Health University
| | - Naomichi Ogitsu
- Department of Clinical Nutrition, Faculty of Health Science, Suzuka University of Medical Science
| | - Misao Yoneda
- Department of Clinical Nutrition, Faculty of Health Science, Suzuka University of Medical Science
| | - Toshihiro Miura
- Department of Clinical Nutrition, Faculty of Health Science, Suzuka University of Medical Science
| | - Yoichi Nagamura
- Department of Clinical Nutrition, Faculty of Health Science, Suzuka University of Medical Science
| | - Shizuko Nagao
- Education and Research Center of Animal Models for Human Diseases, Fujita Health University
| |
Collapse
|
35
|
Ullah R, Rauf N, Nabi G, Yi S, Yu-Dong Z, Fu J. Mechanistic insight into high-fat diet-induced metabolic inflammation in the arcuate nucleus of the hypothalamus. Biomed Pharmacother 2021; 142:112012. [PMID: 34388531 DOI: 10.1016/j.biopha.2021.112012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/31/2021] [Accepted: 08/03/2021] [Indexed: 12/16/2022] Open
Abstract
A high-fat diet (HFD) is linked with cytokines production by non-neuronal cells within the hypothalamus, which mediates metabolic inflammation. These cytokines then activate different inflammatory mediators in the arcuate nucleus of the hypothalamus (ARC), a primary hypothalamic area accommodating proopiomelanocortin (POMC) and agouti-related peptide (AGRP) neurons, first-order neurons that sense and integrate peripheral metabolic signals and then respond accordingly. These mediators, such as inhibitor of κB kinase-β (IKKβ), suppression of cytokine signaling 3 (SOCS3), c-Jun N-terminal kinases (JNKs), protein kinase C (PKC), etc., cause insulin and leptin resistance in POMC and AGRP neurons and support obesity and related metabolic complications. On the other hand, inhibition of these mediators has been shown to counteract the impaired metabolism. Therefore, it is important to discuss the contribution of neuronal and non-neuronal cells in HFD-induced hypothalamic inflammation. Furthermore, understanding few other questions, such as the diets causing hypothalamic inflammation, the gender disparity in response to HFD feeding, and how hypothalamic inflammation affects ARC neurons to cause impaired metabolism, will be helpful for the development of therapeutic approaches to prevent or treat HFD-induced obesity.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China; Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Naveed Rauf
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Ghulam Nabi
- Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China; Department of Life Sciences, School of Science, University of Management and Technology (UMT), Lahore, Pakistan
| | - Shen Yi
- Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
| | - Zhou Yu-Dong
- Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
| | - Junfen Fu
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China; National Clinical Research Center for Child Health, Hangzhou 310052, China; National Children's Regional Medical Center, Hangzhou 310052, China.
| |
Collapse
|
36
|
Wali JA, Solon-Biet SM, Freire T, Brandon AE. Macronutrient Determinants of Obesity, Insulin Resistance and Metabolic Health. BIOLOGY 2021; 10:336. [PMID: 33923531 PMCID: PMC8072595 DOI: 10.3390/biology10040336] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/07/2021] [Indexed: 01/18/2023]
Abstract
Obesity caused by the overconsumption of calories has increased to epidemic proportions. Insulin resistance is often associated with an increased adiposity and is a precipitating factor in the development of cardiovascular disease, type 2 diabetes, and altered metabolic health. Of the various factors contributing to metabolic impairments, nutrition is the major modifiable factor that can be targeted to counter the rising prevalence of obesity and metabolic diseases. However, the macronutrient composition of a nutritionally balanced "healthy diet" are unclear, and so far, no tested dietary intervention has been successful in achieving long-term compliance and reductions in body weight and associated beneficial health outcomes. In the current review, we briefly describe the role of the three major macronutrients, carbohydrates, fats, and proteins, and their role in metabolic health, and provide mechanistic insights. We also discuss how an integrated multi-dimensional approach to nutritional science could help in reconciling apparently conflicting findings.
Collapse
Affiliation(s)
- Jibran A Wali
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
- School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, NSW 2006, Australia
| | - Samantha M Solon-Biet
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Therese Freire
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Amanda E Brandon
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
37
|
Sandovici I, Hammerle CM, Virtue S, Vivas-Garcia Y, Izquierdo-Lahuerta A, Ozanne SE, Vidal-Puig A, Medina-Gómez G, Constância M. Autocrine IGF2 programmes β-cell plasticity under conditions of increased metabolic demand. Sci Rep 2021; 11:7717. [PMID: 33833312 PMCID: PMC8032793 DOI: 10.1038/s41598-021-87292-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/08/2021] [Indexed: 02/07/2023] Open
Abstract
When exposed to nutrient excess and insulin resistance, pancreatic β-cells undergo adaptive changes in order to maintain glucose homeostasis. The role that growth control genes, highly expressed in early pancreas development, might exert in programming β-cell plasticity in later life is a poorly studied area. The imprinted Igf2 (insulin-like growth factor 2) gene is highly transcribed during early life and has been identified in recent genome-wide association studies as a type 2 diabetes susceptibility gene in humans. Hence, here we investigate the long-term phenotypic metabolic consequences of conditional Igf2 deletion in pancreatic β-cells (Igf2βKO) in mice. We show that autocrine actions of IGF2 are not critical for β-cell development, or for the early post-natal wave of β-cell remodelling. Additionally, adult Igf2βKO mice maintain glucose homeostasis when fed a chow diet. However, pregnant Igf2βKO females become hyperglycemic and hyperinsulinemic, and their conceptuses exhibit hyperinsulinemia and placentomegalia. Insulin resistance induced by congenital leptin deficiency also renders Igf2βKO females more hyperglycaemic compared to leptin-deficient controls. Upon high-fat diet feeding, Igf2βKO females are less susceptible to develop insulin resistance. Based on these findings, we conclude that in female mice, autocrine actions of β-cell IGF2 during early development determine their adaptive capacity in adult life.
Collapse
Affiliation(s)
- Ionel Sandovici
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK.
- Department of Obstetrics and Gynaecology and National Institute for Health Research, Cambridge Biomedical Research Centre, Cambridge, CB2 0SW, UK.
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK.
| | - Constanze M Hammerle
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK.
- Department of Obstetrics and Gynaecology and National Institute for Health Research, Cambridge Biomedical Research Centre, Cambridge, CB2 0SW, UK.
- Novo Nordisk A/S, 2880, Bagsværd, Denmark.
| | - Sam Virtue
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Yurena Vivas-Garcia
- Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922, Alcorcón, Madrid, Spain
- Nuffield Department of Clinical Medicine, Ludwig Institute for Cancer Research, University of Oxford, Headington, Oxford, OX3 7DQ, UK
| | - Adriana Izquierdo-Lahuerta
- Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922, Alcorcón, Madrid, Spain
| | - Susan E Ozanne
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Antonio Vidal-Puig
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
- Welcome Trust Sanger Institute, Hinxton, CB10 1SA, UK
- Cambridge University Nanjing Centre of Technology and Innovation, Jiangbei Area, Nanjing, People's Republic of China
| | - Gema Medina-Gómez
- Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922, Alcorcón, Madrid, Spain
| | - Miguel Constância
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK.
- Department of Obstetrics and Gynaecology and National Institute for Health Research, Cambridge Biomedical Research Centre, Cambridge, CB2 0SW, UK.
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK.
| |
Collapse
|
38
|
Integrative analysis of physiological responses to high fat feeding with diffusion tensor images and neurochemical profiles of the mouse brain. Int J Obes (Lond) 2021; 45:1203-1214. [PMID: 33574566 PMCID: PMC8159736 DOI: 10.1038/s41366-021-00775-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/28/2020] [Accepted: 01/21/2021] [Indexed: 02/03/2023]
Abstract
Background Obesity proceeds with important physiological and microstructural alterations in the brain, but the precise relationships between the diet and feeding status, its physiological responses, and the observed neuroimaging repercussions, remain elusive. Here, we implemented a mouse model of high fat diet (HFD) feeding to explore specific associations between diet, feeding status, phenotypic and endocrine repercussions, and the resulting microstructural and metabolic alterations in the brain, as detected by diffusion tensor imaging (DTI) and neurochemical metabolic profiling. Methods Brain DTI images were acquired from adult male C57BL6/J mice after 6 weeks of HFD, or standard diet (SD) administrations, both under the fed, and overnight fasted conditions. Metabolomic profiles of the cortex (Ctx), hippocampus (Hipc), and hypothalamus (Hyp) were determined by 1H high-resolution magic angle spinning (HRMAS) spectroscopy, in cerebral biopsies dissected after microwave fixation. Mean diffusivity (MD), fractional anisotropy (FA) maps, and HRMAS profiles were complemented with determinations of phenotypic alterations and plasma levels of appetite-related hormones, measured by indirect calorimetry and multiplex assays, respectively. We used Z-score and alternating least squares scaling (ALSCAL) analysis to investigate specific associations between diet and feeding status, physiological, and imaging parameters. Results HFD induced significant increases in body weight and the plasma levels of glucose and fatty acids in the fed and fasted conditions, as well as higher cerebral MD (Ctx, Hipc, Hyp), FA (Hipc), and mobile saturated fatty acids resonances (Ctx, Hipc, Hyp). Z-score and ASLCAL analysis identified the precise associations between physiological and imaging variables. Conclusions The present study reveals that diet and feeding conditions elicit prominent effects on specific imaging and spectroscopic parameters of the mouse brain that can be associated to the alterations in phenotypic and endocrine variables. Together, present results disclose a neuro-inflammatory response to HFD, characterized primarily by vasogenic edema and compensatory responses in osmolyte concentrations.
Collapse
|
39
|
Casimiro I, Stull ND, Tersey SA, Mirmira RG. Phenotypic sexual dimorphism in response to dietary fat manipulation in C57BL/6J mice. J Diabetes Complications 2021; 35:107795. [PMID: 33308894 PMCID: PMC7856196 DOI: 10.1016/j.jdiacomp.2020.107795] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/17/2020] [Accepted: 11/07/2020] [Indexed: 01/23/2023]
Abstract
BACKGROUND Obesity and the metabolic syndrome are increasingly prevalent in society and their complications and response to treatment exhibit sexual dimorphism. Mouse models of high fat diet-induced obesity are commonly used for both mechanistic and therapeutic studies of metabolic disease and diabetes. However, the inclusion of female mammals in obesity research has not been a common practice, and has resulted in a paucity of data regarding the effect of sex on metabolic parameters and its applicability to humans. METHODS Here we analyzed male and female C57BL/6 J mice beginning at 4 weeks of age that were placed on a low-fat diet (LFD, 10% calories from fat), a Western Diet (WD, 45% calories from fat), or a high fat diet (HFD, 60% calories from fat). Assessments of body composition, glucose homeostasis, insulin production, and energy metabolism, as well as histological analyses of pancreata were performed. RESULTS Both male and female C57BL/6 J mice had similar increases in total percent body weight gain with both WD and HFD compared to LFD, however, male mice gained weight earlier upon HFD or WD feeding compared to female mice. Male mice maintained their caloric food intake while reducing their locomotor activity with either WD or HFD compared to LFD, whereas female mice increased their caloric food intake with WD feeding. Locomotor activity of female mice did not significantly change upon WD or HFD feeding, yet female mice exhibited increased energy expenditure compared to WD or HFD fed male mice. Glucose tolerance tests performed at 4, 12 and 20 weeks of dietary intervention revealed impaired glucose tolerance that was worse in male mice compared to females. Furthermore, male mice exhibited an increase in pancreatic β cell area as well as reduced insulin sensitivity after HFD feeding compared to WD or LFD, whereas female mice did not. CONCLUSIONS Male and female C57BL/6 J mice exhibited strikingly different responses in weight, food consumption, locomotor activity, energy expenditure and β cell adaptation upon dietary manipulation, with the latter exhibiting less striking phenotypic changes. We conclude that the nature of these responses emphasizes the need to contextualize studies of obesity pathophysiology and treatment with respect to sex.
Collapse
Affiliation(s)
- Isabel Casimiro
- Department of Medicine, Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, IL 60637, United States of America
| | - Natalie D Stull
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, United States of America
| | - Sarah A Tersey
- Department of Medicine, Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, IL 60637, United States of America.
| | - Raghavendra G Mirmira
- Department of Medicine, Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, IL 60637, United States of America.
| |
Collapse
|
40
|
Lalanza JF, Snoeren EMS. The cafeteria diet: A standardized protocol and its effects on behavior. Neurosci Biobehav Rev 2020; 122:92-119. [PMID: 33309818 DOI: 10.1016/j.neubiorev.2020.11.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022]
Abstract
Obesity is a major health risk, with junk food consumption playing a central role in weight gain, because of its high palatability and high-energy nutrients. The Cafeteria (CAF) diet model for animal experiments consists of the same tasty but unhealthy food products that people eat (e.g. hot dogs and muffins), and considers variety, novelty and secondary food features, such as smell and texture. This model, therefore, mimics human eating patterns better than other models. In this paper, we systematically review studies that have used a CAF diet in behavioral experiments and propose a standardized CAF diet protocol. The proposed diet is ad libitum and voluntary; combines different textures, nutrients and tastes, including salty and sweet products; and it is rotated and varied. Our summary of the behavioral effects of CAF diet show that it alters meal patterns, reduces the hedonic value of other rewards, and tends to reduce stress and spatial memory. So far, no clear effects of CAF diet were found on locomotor activity, impulsivity, coping and social behavior.
Collapse
Affiliation(s)
- Jaume F Lalanza
- Department of Psychology, UiT The Arctic University of Norway, Tromsø, Norway.
| | - Eelke M S Snoeren
- Department of Psychology, UiT The Arctic University of Norway, Tromsø, Norway; Regional Health Authority of North Norway, Norway.
| |
Collapse
|
41
|
Preguiça I, Alves A, Nunes S, Fernandes R, Gomes P, Viana SD, Reis F. Diet-induced rodent models of obesity-related metabolic disorders-A guide to a translational perspective. Obes Rev 2020; 21:e13081. [PMID: 32691524 DOI: 10.1111/obr.13081] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/12/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022]
Abstract
Diet is a critical element determining human health and diseases, and unbalanced food habits are major risk factors for the development of obesity and related metabolic disorders. Despite technological and pharmacological advances, as well as intensification of awareness campaigns, the prevalence of metabolic disorders worldwide is still increasing. Thus, novel therapeutic approaches with increased efficacy are urgently required, which often depends on cellular and molecular investigations using robust animal models. In the absence of perfect rodent models, those induced by excessive consumption of fat and sugars better replicate the key aspects that are the root causes of human metabolic diseases. However, the results obtained using these models cannot be directly compared, particularly because of the use of different dietary protocols, and animal species and strains, among other confounding factors. This review article revisits diet-induced models of obesity and related metabolic disorders, namely, metabolic syndrome, prediabetes, diabetes and nonalcoholic fatty liver disease. A critical analysis focused on the main pathophysiological features of rodent models, as opposed to the criteria defined for humans, is provided as a practical guide with a translational perspective for the establishment of animal models of obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Inês Preguiça
- Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
| | - André Alves
- Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
| | - Sara Nunes
- Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
| | - Rosa Fernandes
- Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
| | - Pedro Gomes
- Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal.,Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Center for Health Technology and Services Research (CINTESIS), University of Porto, Porto, Portugal
| | - Sofia D Viana
- Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal.,ESTESC-Coimbra Health School, Pharmacy, Polytechnic Institute of Coimbra, Coimbra, Portugal
| | - Flávio Reis
- Institute of Pharmacology and Experimental Therapeutics, and Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
42
|
Velázquez AM, Roglans N, Bentanachs R, Gené M, Sala-Vila A, Lázaro I, Rodríguez-Morató J, Sánchez RM, Laguna JC, Alegret M. Effects of a Low Dose of Caffeine Alone or as Part of a Green Coffee Extract, in a Rat Dietary Model of Lean Non-Alcoholic Fatty Liver Disease without Inflammation. Nutrients 2020; 12:nu12113240. [PMID: 33113993 PMCID: PMC7690747 DOI: 10.3390/nu12113240] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/16/2022] Open
Abstract
Non-alcoholic fatty liver disease is a highly prevalent condition without specific pharmacological treatment, characterized in the initial stages by hepatic steatosis. It was suggested that lipid infiltration in the liver might be reduced by caffeine through anti-inflammatory, antioxidative, and fatty acid metabolism-related mechanisms. We investigated the effects of caffeine (CAF) and green coffee extract (GCE) on hepatic lipids in lean female rats with steatosis. For three months, female Sprague-Dawley rats were fed a standard diet or a cocoa butter-based high-fat diet plus 10% liquid fructose. In the last month, the high-fat diet was supplemented or not with CAF or a GCE, providing 5 mg/kg of CAF. Plasma lipid levels and the hepatic expression of molecules involved in lipid metabolism were determined. Lipidomic analysis was performed in liver samples. The diet caused hepatic steatosis without obesity, inflammation, endoplasmic reticulum stress, or hepatic insulin resistance. Neither CAF nor GCE alleviated hepatic steatosis, but GCE-treated rats showed lower hepatic triglyceride levels compared to the CAF group. The GCE effects could be related to reductions of hepatic (i) mTOR phosphorylation, leading to higher nuclear lipin-1 levels and limiting lipogenic gene expression; (ii) diacylglycerol levels; (iii) hexosylceramide/ceramide ratios; and (iv) very-low-density lipoprotein receptor expression. In conclusion, a low dose of CAF did not reduce hepatic steatosis in lean female rats, but the same dose provided as a green coffee extract led to lower liver triglyceride levels.
Collapse
Affiliation(s)
- Ana Magdalena Velázquez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Avda Joan XXIII 27-31, 08028 Barcelona, Spain; (A.M.V.); (N.R.); (R.B.); (M.G.); (R.M.S.)
| | - Núria Roglans
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Avda Joan XXIII 27-31, 08028 Barcelona, Spain; (A.M.V.); (N.R.); (R.B.); (M.G.); (R.M.S.)
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - Roger Bentanachs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Avda Joan XXIII 27-31, 08028 Barcelona, Spain; (A.M.V.); (N.R.); (R.B.); (M.G.); (R.M.S.)
| | - Maria Gené
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Avda Joan XXIII 27-31, 08028 Barcelona, Spain; (A.M.V.); (N.R.); (R.B.); (M.G.); (R.M.S.)
| | - Aleix Sala-Vila
- IMIM-Hospital del Mar Medical Research Institute, 08003 Barcelona, Spain; (A.S.-V.); (I.L.)
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, 08005 Barcelona, Spain
| | - Iolanda Lázaro
- IMIM-Hospital del Mar Medical Research Institute, 08003 Barcelona, Spain; (A.S.-V.); (I.L.)
| | - Jose Rodríguez-Morató
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
- IMIM-Hospital del Mar Medical Research Institute, 08003 Barcelona, Spain; (A.S.-V.); (I.L.)
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra (CEXS-UPF), 08003 Barcelona, Spain
| | - Rosa María Sánchez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Avda Joan XXIII 27-31, 08028 Barcelona, Spain; (A.M.V.); (N.R.); (R.B.); (M.G.); (R.M.S.)
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - Juan Carlos Laguna
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Avda Joan XXIII 27-31, 08028 Barcelona, Spain; (A.M.V.); (N.R.); (R.B.); (M.G.); (R.M.S.)
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
- Correspondence: (J.C.L.); (M.A.); Tel.: +34-93-4024531 (M.A.)
| | - Marta Alegret
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Avda Joan XXIII 27-31, 08028 Barcelona, Spain; (A.M.V.); (N.R.); (R.B.); (M.G.); (R.M.S.)
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
- Correspondence: (J.C.L.); (M.A.); Tel.: +34-93-4024531 (M.A.)
| |
Collapse
|
43
|
Wali JA, Jarzebska N, Raubenheimer D, Simpson SJ, Rodionov RN, O’Sullivan JF. Cardio-Metabolic Effects of High-Fat Diets and Their Underlying Mechanisms-A Narrative Review. Nutrients 2020; 12:E1505. [PMID: 32455838 PMCID: PMC7284903 DOI: 10.3390/nu12051505] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
The majority of the epidemiological evidence over the past few decades has linked high intake of fats, especially saturated fats, to increased risk of diabetes and cardiovascular disease. However, findings of some recent studies (e.g., the PURE study) have contested this association. High saturated fat diets (HFD) have been widely used in rodent research to study the mechanism of insulin resistance and metabolic syndrome. Two separate but somewhat overlapping models-the diacylglycerol (DAG) model and the ceramide model-have emerged to explain the development of insulin resistance. Studies have shown that lipid deposition in tissues such as muscle and liver inhibit insulin signaling via the toxic molecules DAG and ceramide. DAGs activate protein kinase C that inhibit insulin-PI3K-Akt signaling by phosphorylating serine residues on insulin receptor substrate (IRS). Ceramides are sphingolipids with variable acyl group chain length and activate protein phosphatase 2A that dephosphorylates Akt to block insulin signaling. In adipose tissue, obesity leads to infiltration of macrophages that secrete pro-inflammatory cytokines that inhibit insulin signaling by phosphorylating serine residues of IRS proteins. For cardiovascular disease, studies in humans in the 1950s and 1960s linked high saturated fat intake with atherosclerosis and coronary artery disease. More recently, trials involving Mediterranean diet (e.g., PREDIMED study) have indicated that healthy monounsaturated fats are more effective in preventing cardiovascular mortality and coronary artery disease than are low-fat, low-cholesterol diets. Antioxidant and anti-inflammatory effects of Mediterranean diets are potential mediators of these benefits.
Collapse
Affiliation(s)
- Jibran A. Wali
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (D.R.); (S.J.S.)
- Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Natalia Jarzebska
- University Center for Vascular Medicine Department of Medicine III—Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (N.J.); (R.N.R.)
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - David Raubenheimer
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (D.R.); (S.J.S.)
- Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Stephen J. Simpson
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (D.R.); (S.J.S.)
- Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Roman N. Rodionov
- University Center for Vascular Medicine Department of Medicine III—Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (N.J.); (R.N.R.)
| | - John F. O’Sullivan
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; (D.R.); (S.J.S.)
- Faculty of Medical Sciences, School of Medicine, The University of Sydney, Sydney, NSW 2006, Australia
- Heart Research Institute, The University of Sydney, Sydney, NSW 2006, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| |
Collapse
|
44
|
Hubbard K, Shome A, Sun B, Pontré B, McGregor A, Mountjoy KG. Chronic High-Fat Diet Exacerbates Sexually Dimorphic Pomctm1/tm1 Mouse Obesity. Endocrinology 2019; 160:1081-1096. [PMID: 30997487 PMCID: PMC6469954 DOI: 10.1210/en.2018-00924] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/04/2019] [Indexed: 01/16/2023]
Abstract
Mice with a targeted mutation in the pro-opiomelanocortin (Pomc) gene (Pomctm1/tm1 mice) are unable to synthesize desacetyl-α-MSH and α-MSH and they develop obesity when fed chow diet. In this study, we hypothesized that a chronic high-fat (HF) diet exacerbates Pomctm1/tm1 mouse obesity. Male and female Pomcwt/wt and Pomctm1/tm1 mice were fed low-fat (LF) (10 kcal percent fat) or HF (45 kcal percent fat) diets from weaning for 23 weeks. We show that Pomctm1/tm1 mouse obesity is sexually dimorphic and exacerbated by an HF diet. Male Pomctm1/tm1 mice develop obesity because they are hyperphagic compared with Pomcwt/wt mice when fed an LF or HF diet. Female Pomctm1/tm1 mice develop obesity when feeding on an LF or HF diet because they exhibit signs of reduced energy expenditure (no change in feed efficiency; body weight gained exceeding energy intake) compared with Pomcwt/wt mice. A chronic HF diet exacerbates male Pomctm1/tm1 and Pomcwt/wt mouse obesity, and the increased energy intake fully accounts for increased weight gain. In contrast, female Pomcwt/wt mice are protected from chronic HF diet-induced obesity because they reduce the amount of HF diet eaten, and they appear to increase their energy expenditure (no change in feed efficiency but energy intake exceeding body weight gained). A chronic HF diet exacerbates female Pomctm1/tm1 mouse obesity due to impaired ability to reduce the amount of HF diet eaten and apparent impaired HF diet-induced adaptive thermogenesis. Our data show that desacetyl-α-MSH and α-MSH are required for sexually dimorphic HF diet-induced C57BL/6J obesity. In conclusion, desacetyl-α-MSH and α-MSH play salutary roles in sexually dimorphic melanocortin obesity and sexually dimorphic HF diet-induced C57BL/6J obesity.
Collapse
Affiliation(s)
- Kristina Hubbard
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Avik Shome
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Bo Sun
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Beau Pontré
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Ailsa McGregor
- Department of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kathleen G Mountjoy
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Biodiscovery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
45
|
Purnell JQ, Urbanski HF, Kievit P, Roberts CT, Bethea CL. Estradiol Replacement Timing and Obesogenic Diet Effects on Body Composition and Metabolism in Postmenopausal Macaques. Endocrinology 2019; 160:899-914. [PMID: 30753523 PMCID: PMC6435013 DOI: 10.1210/en.2018-00884] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/02/2019] [Indexed: 12/14/2022]
Abstract
Whether hormone replacement therapy has beneficial metabolic effects in postmenopausal women remains controversial because of between-study differences in menopausal duration, estrogen formulations, and diet. Additionally, animal studies have not reflected the typical human obesogenic, Western-style diet (WSD). In this study, we determined the effects of immediate 17β-estradiol (ImE) or delayed 17β-estradiol treatment on weight and metabolism parameters in old ovo-hysterectomized rhesus macaques consuming a WSD over a 30-month period. The placebo and ImE groups exhibited progressive gains in weight and fat mass, which ImE initially attenuated but did not prevent. Progression of insulin resistance (IR) was lessened by ImE compared with placebo under both fasting and IV glucose-stimulated conditions, plateauing in all groups between 24 and 30 months. Consequently, relative euglycemia was maintained through lower stimulated insulin levels with ImE than with placebo. Bone mineral density decreased in the placebo group but was maintained in the ImE group, whereas bone mineral content was unaffected by placebo and increased with ImE. Daily activity was reduced while macaques consumed a WSD and was not affected by ImE. Over time, total cholesterol, triglyceride, very-low-density cholesterol, high-density lipoprotein cholesterol (HDL-C), non-HDL-C, and IL-8 levels increased or trended upward in all animals, with only the change in HDL-C affected by ImE. Delayed estrogen treatment (months 24 to 30) had no significant impact on body composition or glucometabolic parameters. In summary, detrimental WSD-induced changes in body composition and metabolism were only temporarily ameliorated by ImE, with the important exception of glucose homeostasis, which benefited from E replacement even as body composition worsened.
Collapse
Affiliation(s)
- Jonathan Q Purnell
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
- Division of Endocrinology, Diabetes, Clinical Nutrition, Oregon Health & Science University, Portland, Oregon
- Correspondence: Jonathan Q. Purnell, MD, Oregon Health & Science University, Mailstop MDYMI, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239. E-mail:
| | - Henryk F Urbanski
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon
| | - Charles T Roberts
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon
| | - Cynthia L Bethea
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
46
|
Brunetta HS, de Paula GC, de Oliveira J, Martins EL, Dos Santos GJ, Galina A, Rafacho A, de Bem AF, Nunes EA. Decrement in resting and insulin-stimulated soleus muscle mitochondrial respiration is an early event in diet-induced obesity in mice. Exp Physiol 2019; 104:306-321. [PMID: 30578638 DOI: 10.1113/ep087317] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 12/19/2018] [Indexed: 01/08/2023]
Abstract
NEW FINDINGS What is the central question of this study? What are the temporal responses of mitochondrial respiration and mitochondrial responsivity to insulin in soleus muscle fibres from mice during the development of obesity and insulin resistance? What is the main finding and its importance? Short- and long-term feeding with a high-fat diet markedly reduced soleus mitochondrial respiration and mitochondrial responsivity to insulin before any change in glycogen synthesis. Muscle glycogen synthesis and whole-body insulin resistance were present after 14 and 28 days, respectively. Our findings highlight the plasticity of mitochondria during the development of obesity and insulin resistance. ABSTRACT Recently, significant attention has been given to the role of muscle mitochondrial function in the development of insulin resistance associated with obesity. Our aim was to investigate temporal alterations in mitochondrial respiration, H2 O2 emission and mitochondrial responsivity to insulin in permeabilized skeletal muscle fibres during the development of obesity in mice. Male Swiss mice (5-6 weeks old) were fed with a high-fat diet (60% calories from fat) or standard diet for 7, 14 or 28 days to induce obesity and insulin resistance. Diet-induced obese (DIO) mice presented with reduced glucose tolerance and hyperinsulinaemia after 7 days of high-fat diet. After 14 days, the expected increase in muscle glycogen content after systemic injection of glucose and insulin was not observed in DIO mice. At 28 days, blood glucose decay after insulin injection was significantly impaired. Complex I (pyruvate + malate) and II (succinate)-linked respiration and oxidative phosphorylation (ADP) were decreased after 7 days of high-fat diet and remained low in DIO mice after 14 and 28 days of treatment. Moreover, mitochondria from DIO mice were incapable of increasing respiratory coupling and ADP responsivity after insulin stimulation in all observed periods. Markers of mitochondrial content were reduced only after 28 days of treatment. The mitochondrial H2 O2 emission profile varied during the time course of DIO, with a reduction of H2 O2 emission in the early stages of DIO and an increased emission after 28 days of treatment. Our data demonstrate that DIO promotes transitory alterations in mitochondrial physiology during the early and late stages of insulin resistance related to obesity.
Collapse
Affiliation(s)
- Henver Simionato Brunetta
- Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina, Florianopólis, Santa Catrina, Brazil
| | - Gabriela Cristina de Paula
- Graduate Program in Biochemistry, Federal University of Santa Catarina, Florianopólis, Santa Catrina, Brazil
| | - Jade de Oliveira
- Graduate Program in Health Sciences, University of Extremo Sul Catarinense, Criciúma, Santa Catarina, Brazil
| | - Eduarda Lopes Martins
- Graduate Program in Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Jorge Dos Santos
- Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina, Florianopólis, Santa Catrina, Brazil
| | - Antonio Galina
- Graduate Program in Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alex Rafacho
- Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina, Florianopólis, Santa Catrina, Brazil
| | - Andreza Fabro de Bem
- Graduate Program in Biochemistry, Federal University of Santa Catarina, Florianopólis, Santa Catrina, Brazil.,Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Distrito Federal, Brazil
| | - Everson Araújo Nunes
- Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina, Florianopólis, Santa Catrina, Brazil
| |
Collapse
|
47
|
Kuda O. On the Complexity of PAHSA Research. Cell Metab 2018; 28:541-542. [PMID: 30244970 DOI: 10.1016/j.cmet.2018.09.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 01/28/2023]
Abstract
Reporting in Cell Metabolism, Pflimlin et al. recently found no beneficial effect of PAHSAs on glucose control in mice on several high-fat diets. Kuda cautions that due to methodological differences, the data must be carefully reinterpreted, emphasizing that olive oil contains high amounts of FAHFAs, potentially masking the effects of PAHSAs in studies utilizing this vehicle.
Collapse
Affiliation(s)
- Ondrej Kuda
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic.
| |
Collapse
|
48
|
|
49
|
Wang S, Wang C, Turdi S, Richmond KL, Zhang Y, Ren J. ALDH2 protects against high fat diet-induced obesity cardiomyopathy and defective autophagy: role of CaM kinase II, histone H3K9 methyltransferase SUV39H, Sirt1, and PGC-1α deacetylation. Int J Obes (Lond) 2018. [PMID: 29535452 DOI: 10.1038/s41366-018-0030-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Uncorrected obesity contributes to cardiac remodeling and contractile dysfunction although the underlying mechanism remains poorly understood. Mitochondrial aldehyde dehydrogenase (ALDH2) is a mitochondrial enzyme with some promises in a number of cardiovascular diseases. This study was designed to evaluate the impact of ALDH2 on cardiac remodeling and contractile property in high fat diet-induced obesity. METHODS Wild-type (WT) and ALDH2 transgenic mice were fed low (10% calorie from fat) or high (45% calorie from fat) fat diet for 5 months prior to the assessment of cardiac geometry and function using echocardiography, IonOptix system, Lectin, and Masson Trichrome staining. Western blot analysis was employed to evaluate autophagy, CaM kinase II, PGC-1α, histone H3K9 methyltransferase SUV39H, and Sirt-1. RESULTS Our data revealed that high fat diet intake promoted weight gain, cardiac remodeling (hypertrophy and interstitial fibrosis, p < 0.0001) and contractile dysfunction (reduced fractional shortening (p < 0.0001), cardiomyocyte function (p < 0.0001), and intracellular Ca2+ handling (p = 0.0346)), mitochondrial injury (elevated O2- levels, suppressed PGC-1α, and enhanced PGC-1α acetylation, p < 0.0001), elevated SUV39H, suppressed Sirt1, autophagy and phosphorylation of AMPK and CaM kinase II, the effects of which were negated by ALDH2 (p ≤ 0.0162). In vitro incubation of the ALDH2 activator Alda-1 rescued against palmitic acid-induced changes in cardiomyocyte function, the effect of which was nullified by the Sirt-1 inhibitor nicotinamide and the CaM kinase II inhibitor KN-93 (p < 0.0001). The SUV39H inhibitor chaetocin mimicked Alda-1-induced protection again palmitic acid (p < 0.0001). Examination in overweight human revealed an inverse correlation between diastolic cardiac function and ALDH2 gene mutation (p < 0.05). CONCLUSIONS Taken together, these data suggest that ALDH2 serves as an indispensable factor against cardiac anomalies in diet-induced obesity through a mechanism related to autophagy regulation and facilitation of the SUV39H-Sirt1-dependent PGC-1α deacetylation.
Collapse
Affiliation(s)
- Shuyi Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.,Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA
| | - Cong Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Subat Turdi
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA
| | - Kacy L Richmond
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA
| | - Yingmei Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China. .,Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China. .,Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA.
| |
Collapse
|
50
|
Tucker LA. Fiber Intake and Insulin Resistance in 6374 Adults: The Role of Abdominal Obesity. Nutrients 2018; 10:E237. [PMID: 29461482 PMCID: PMC5852813 DOI: 10.3390/nu10020237] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 01/02/2023] Open
Abstract
A cross-sectional design was used to evaluate the relationship between fiber intake and insulin resistance, indexed using HOMA (homeostatic model assessment), in a National Health and Nutrition Examination Study (NHANES) sample of 6374 U.S. adults. Another purpose was to test the influence of covariates on the association. A third aim was to compare HOMA levels between two groups based on the recommended intake of 14 grams of fiber per 1000 kilocalories (kcal). Fiber intake was measured using a 24-hour recall. With demographic variables controlled, results showed that HOMA differed across High, Moderate, and Low fiber categories (F = 5.4, p = 0.0072). Adjusting for the demographic variables, the possible misreporting of energy intake, smoking, and physical activity strengthened the relationship (F = 8.0, p = 0.0009), which remained significant after adjusting for body fat (F = 7.0, p = 0.0019) and body mass index (BMI) (F = 4.9, p = 0.0108), with the other covariates. However, the fiber-HOMA relationship was eliminated after adjusting for waist circumference (F = 2.3, p = 0.1050). Dividing participants based on the recommended 14-gram standard resulted in meaningful HOMA differences (F = 16.4, p = 0.0002), and the association was not eliminated after controlling for waist circumference. Apparently, adults with high fiber consumption have less insulin resistance than their counterparts. However, much of the association is due to differences in waist circumference, unless the recommended intake of fiber is attained.
Collapse
Affiliation(s)
- Larry A Tucker
- Department of Exercise Sciences, Brigham Young University, Provo 84602, UT, USA.
| |
Collapse
|