1
|
Aggarwal H, Gautam J, Gupta SK, Das B, Kumar Y, Jagavelu K, Dikshit M. Improved metabolic stability in iNOS knockout mice with Lactobacillus supplementation. Nutr Res 2024; 132:95-111. [PMID: 39532058 DOI: 10.1016/j.nutres.2024.09.018] [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: 02/21/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024]
Abstract
Oxidative and nitrosative stress play pivotal roles in normal physiological processes and the pathogenesis of metabolic disorders. Previous studies from our lab demonstrated insulin resistance (IR), and dyslipidemia in iNOS-/- mice, emphasizing the importance of maintaining optimal redox balance. These mice exhibited altered gut microbiota with decreased Lactobacillus. Therefore, we hypothesized that Lactobacillus supplementation could mitigate metabolic disturbances in iNOS-/- mice. To test this hypothesis, iNOS-/- mice and wild-type (WT) mice were divided into four groups: iNOS-/- with or without Lactobacillus supplementation, WT with or without Lactobacillus supplementation and glucose tolerance, insulin resistance, gluconeogenesis, lipids, gene expression related to glucose and lipid metabolism (qPCR), fecal gut microbiota (16S rRNA sequencing), and serum and caecum metabolomics (LC-MS) were monitored. IR and dyslipidemic iNOS-/- mice exhibited reduced microbial diversity, diminished presence of Lactobacillus, and altered serum metabolites, indicating metabolic dysregulation. Lactobacillus supplementation in iNOS-/- mice effectively reversed glucose intolerance, IR, dyslipidemia, and associated metabolic irregularities compared to WT. These improvements correlated with changes in gene expression related to fatty acid synthesis in liver and adipose tissue, lipid oxidation in liver, and lipid efflux in intestinal tissue as compared to untreated iNOS-/- mice. Despite the positive effects on metabolic markers, Lactobacillus supplementation did not reduce body weight or rectify disrupted energy balance, as evidenced by reduced VCO2 production, heat generation, and metabolic rates in iNOS-/- mice. The results suggest that Lactobacillus supplementation ameliorates metabolic disturbances but did not fully restore disrupted energy balance, highlighting complex interactions between the gut microbiome and metabolism.
Collapse
Affiliation(s)
- Hobby Aggarwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India; Non-communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India
| | - Jyoti Gautam
- Non-communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India; Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Sonu Kumar Gupta
- Non-communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India
| | - Bhabatosh Das
- Molecular Genetics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India
| | - Yashwant Kumar
- Non-communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India
| | - Kumaravelu Jagavelu
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
| | - Madhu Dikshit
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India; Non-communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India.
| |
Collapse
|
2
|
Anwar A, Shukla S, Pathak P. Nitric oxide in modulating oxidative stress mediated skeletal muscle insulin resistance. Mol Biol Rep 2024; 51:944. [PMID: 39210004 DOI: 10.1007/s11033-024-09874-y] [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: 05/06/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Insulin resistance (IR) being the major cause behind different metabolic disorders, has attracted a lot of attention. Epidemiological data shows marked rise in the cases over a period of time. Nitric oxide (NO), produced from nitric oxide synthases (NOS), is involved in a variety of biological functions, alteration in which causes various disorders like hypertension, atherosclerosis, and angiogenesis-associated disorders. IR has been found to be a contributing factor, which is associated with abnormal NO signalling. Skeletal muscle is essential for metabolism, both for its role in glucose uptake and its importance in metabolic disease. In this article, we give an overview of the significance of NO in oxidative stress (OS) mediated IR, describing its role in different conditions that are associated with skeletal muscle IR. NO is found to be involved in the activation of insulin receptor downstream pathway, which suggests absence of NO could lead to reduced glucose uptake, and may ultimately result in IR.
Collapse
Affiliation(s)
- Aamir Anwar
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University (Lucknow Campus), Lucknow, Uttar Pradesh, 226010, India
| | - Shivang Shukla
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University (Lucknow Campus), Lucknow, Uttar Pradesh, 226010, India
| | - Priya Pathak
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University (Lucknow Campus), Lucknow, Uttar Pradesh, 226010, India.
| |
Collapse
|
3
|
Sáez T, Pageé A, Kirschenman R, Quon A, Spaans F, Davidge ST. A High Cholesterol Diet During Late Pregnancy Impairs Long-Term Maternal Vascular Function in Mice. Arterioscler Thromb Vasc Biol 2023; 43:120-132. [PMID: 36353990 DOI: 10.1161/atvbaha.122.318421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Gestational dyslipidemia is associated with pregnancy complications including preeclampsia. However, whether gestational dyslipidemia leads postpartum vascular dysfunction, which could increase the risk for cardiovascular complications later in life, is not known. Here, we aimed to determine whether a gestational dyslipidemia affects postpartum vascular health and induces early signs of atherosclerosis. METHODS Pregnant C57BL/6 mice received a high cholesterol diet or control diet from gestational day 13.5 until term. After delivery, all mice received the control diet for ≈3 months postpartum (PP). Age-matched nulliparous females were on the same diets for equal periods. After 3 months, all mice were euthanized, serum was collected, and aortas were isolated to assess vascular function (wire myography) and markers of oxidative stress and early atherosclerosis. RESULTS PP-high cholesterol diet females had increased circulating cholesterol levels compared with PP-control diet mice, without effect of the diet in nulliparous mice. Methacholine-induced vasodilation was impaired, and nitric oxide contribution reduced, by the high cholesterol diet in aortas of PP mice, but not in nulliparous mice. Exposure to oxidized low-density-protein cholesterol further impaired methylcholine-induced vasodilation in PP-high cholesterol diet aortas only. Compared with PP-control diet mice, aortic inducible nitric oxide synthase expression, reactive oxygen species and nitrotyrosine levels were increased in aortas from PP-high cholesterol diet mice. No differences in aortic lipid deposition and macrophage infiltration were found. CONCLUSIONS Exposure to a high cholesterol diet in pregnancy impairs vascular function postpartum. Our results support the hypothesis that gestational dyslipidemia impacts maternal vascular function after pregnancy, which could potentially predispose these women to future cardiovascular complications.
Collapse
Affiliation(s)
- Tamara Sáez
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada (T.S., R.K., A.Q., F.S., S.T.D.).,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada (T.S., R.K., A.Q., F.S., S.T.D.)
| | - Abbey Pageé
- Department of Physiology, University of Alberta, Edmonton, Canada (A.P., S.T.D.)
| | - Raven Kirschenman
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada (T.S., R.K., A.Q., F.S., S.T.D.).,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada (T.S., R.K., A.Q., F.S., S.T.D.)
| | - Anita Quon
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada (T.S., R.K., A.Q., F.S., S.T.D.).,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada (T.S., R.K., A.Q., F.S., S.T.D.)
| | - Floor Spaans
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada (T.S., R.K., A.Q., F.S., S.T.D.).,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada (T.S., R.K., A.Q., F.S., S.T.D.)
| | - Sandra T Davidge
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada (T.S., R.K., A.Q., F.S., S.T.D.).,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada (T.S., R.K., A.Q., F.S., S.T.D.).,Department of Physiology, University of Alberta, Edmonton, Canada (A.P., S.T.D.)
| |
Collapse
|
4
|
Inci N, Akyildiz EO, Bulbul AA, Turanli ET, Akgun E, Baykal AT, Colak F, Bozaykut P. Transcriptomics and Proteomics Analyses Reveal JAK Signaling and Inflammatory Phenotypes during Cellular Senescence in Blind Mole Rats: The Reflections of Superior Biology. BIOLOGY 2022; 11:biology11091253. [PMID: 36138732 PMCID: PMC9495822 DOI: 10.3390/biology11091253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/04/2022] [Accepted: 08/15/2022] [Indexed: 11/28/2022]
Abstract
Simple Summary Blind mole rats (BMR) (Spalax, Nannospalax sp.) are extraordinary organisms with cancer resistance and a long lifespan for their size. Cellular senescence is a condition in which cells cease dividing irreversibly and secrete proinflammatory cytokines. To understand the mechanisms behind their superior traits, we utilized transcriptomics and proteomics tools in senescent BMR cells to compare them to similarly sized mice. The results revealed the alterations in Janus kinase (JAK) signaling and the cytokine-mediated pathway during the cellular senescence process in BMRs. These findings might reveal the novel mechanisms behind the unique biology of BMRs through cytokine-mediated adaptations. Abstract The blind mole rat (BMR), a long-living subterranean rodent, is an exceptional model for both aging and cancer research since they do not display age-related phenotypes or tumor formation. The Janus kinase–signal transducer and activator of transcription (JAK–STAT) signaling is a cytokine-stimulated pathway that has a crucial role in immune regulation, proliferation, and cytokine production. Therefore, the pathway has recently attracted interest in cellular senescence studies. Here, by using publicly available data, we report that JAK–STAT signaling was suppressed in the BMR in comparison to the mouse. Interestingly, our experimental results showed upregulated Jak1/2 expressions in BMR fibroblasts during the replicative senescence process. The transcriptomic analysis using publicly available data also demonstrated that various cytokines related to JAK–STAT signaling were upregulated in the late passage cells, while some other cytokines such as MMPs and SERPINs were downregulated, representing a possible balance of senescence-associated secretory phenotypes (SASPs) in the BMR. Finally, our proteomics data also confirmed cytokine-mediated signaling activation in senescent BMR fibroblasts. Together, our findings suggest the critical role of JAK–STAT and cytokine-mediated signaling pathways during cellular senescence, pointing to the possible contribution of divergent inflammatory factors to the superior resistance of aging and cancer in BMRs.
Collapse
Affiliation(s)
- Nurcan Inci
- Department of Molecular and Translational Biomedicine, Institute of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Erdogan Oguzhan Akyildiz
- Department of Molecular and Translational Biomedicine, Institute of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Abdullah Alper Bulbul
- Department of Biostatistics and Bioinformatics, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Eda Tahir Turanli
- Department of Molecular and Translational Biomedicine, Institute of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Emel Akgun
- Department of Medical Biochemistry, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Ahmet Tarik Baykal
- Department of Medical Biochemistry, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Faruk Colak
- Department of Biology, Faculty of Arts and Science, Zonguldak Bulent Ecevit University, Zonguldak 67100, Turkey
| | - Perinur Bozaykut
- Department of Molecular and Translational Biomedicine, Institute of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
- Correspondence:
| |
Collapse
|
5
|
Aggarwal H, Pathak P, Gupta SK, Kumar Y, Jagavelu K, Dikshit M. Serum and cecal metabolic profile of the insulin resistant and dyslipidemic p47 phox knockout mice. Free Radic Res 2022; 56:483-497. [PMID: 36251883 DOI: 10.1080/10715762.2022.2133705] [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: 02/07/2023]
Abstract
Involvement of NOX-dependent oxidative stress in the pathophysiology of metabolic disorders as well as in the maintenance of metabolic homeostasis has been demonstrated previously. In the present study, the metabolic profile in p47phox-/- and WT mice fed on a chow diet was evaluated to assess the role of metabolites in glucose intolerance and dyslipidemia under altered oxidative stress conditions. p47phox-/- mice displayed glucose intolerance, dyslipidemia, hyperglycemia, insulin resistance (IR), hyperinsulinemia, and altered energy homeostasis without any significant change in gluconeogenesis. The expression of genes involved in lipid synthesis and uptake was enhanced in the liver, adipose tissue, and intestine tissues. Similarly, the expression of genes associated with lipid efflux in the liver and intestine was also enhanced. Enhanced gut permeability, inflammation, and shortening of the gut was evident in p47phox-/- mice. Circulating levels of pyrimidines, phosphatidylglycerol lipids, and 3-methyl-2-oxindole were augmented, while level of purine was reduced in the serum. Moreover, the cecal metabolome was also altered, as was evident with the increase in indole-3-acetamide, N-acetyl galactosamine, glycocholate, and a decrease in hippurate, indoxyl sulfate, and indigestible sugars (raffinose and melezitose). Treatment of p47phox-/- mice with pioglitazone, marginally improved glucose intolerance, and dyslipidemia, with an increase in PUFAs (linoleate, docosahexaenoic acid, and arachidonic acid). Overall, the results obtained in p47phox-/- mice indicate an association of IR and dyslipidemia with altered serum and cecal metabolites (both host and bacterial-derived), implying a critical role of NOX-derived ROS in metabolic homeostasis.
Collapse
Affiliation(s)
- Hobby Aggarwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India.,Non-Communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Priya Pathak
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sonu Kumar Gupta
- Non-Communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Yashwant Kumar
- Non-Communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, India
| | | | - Madhu Dikshit
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India.,Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
6
|
Aggarwal H, Pathak P, Singh V, Kumar Y, Shankar M, Das B, Jagavelu K, Dikshit M. Vancomycin-Induced Modulation of Gram-Positive Gut Bacteria and Metabolites Remediates Insulin Resistance in iNOS Knockout Mice. Front Cell Infect Microbiol 2022; 11:795333. [PMID: 35127558 PMCID: PMC8807491 DOI: 10.3389/fcimb.2021.795333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/23/2021] [Indexed: 12/27/2022] Open
Abstract
The role of oxidative and nitrosative stress has been implied in both physiology and pathophysiology of metabolic disorders. Inducible nitric oxide synthase (iNOS) has emerged as a crucial regulator of host metabolism and gut microbiota activity. The present study examines the role of the gut microbiome in determining host metabolic functions in the absence of iNOS. Insulin-resistant and dyslipidemic iNOS-/- mice displayed reduced microbial diversity, with a higher relative abundance of Allobaculum and Bifidobacterium, gram-positive bacteria, and altered serum metabolites along with metabolic dysregulation. Vancomycin, which largely depletes gram-positive bacteria, reversed the insulin resistance (IR), dyslipidemia, and related metabolic anomalies in iNOS-/- mice. Such improvements in metabolic markers were accompanied by alterations in the expression of genes involved in fatty acid synthesis in the liver and adipose tissue, lipid uptake in adipose tissue, and lipid efflux in the liver and intestine tissue. The rescue of IR in vancomycin-treated iNOS-/- mice was accompanied with the changes in select serum metabolites such as 10-hydroxydecanoate, indole-3-ethanol, allantoin, hippurate, sebacic acid, aminoadipate, and ophthalmate, along with improvement in phosphatidylethanolamine to phosphatidylcholine (PE/PC) ratio. In the present study, we demonstrate that vancomycin-mediated depletion of gram-positive bacteria in iNOS-/- mice reversed the metabolic perturbations, dyslipidemia, and insulin resistance.
Collapse
Affiliation(s)
- Hobby Aggarwal
- Pharmacology Division, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, India
| | - Priya Pathak
- Pharmacology Division, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, India
| | - Vishal Singh
- Department of Nutritional Sciences, The Pennsylvania State University, State College, PA, United States
| | - Yashwant Kumar
- Non-Communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Manoharan Shankar
- Microbial Physiology Laboratory, Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, India
| | - Bhabatosh Das
- Molecular Genetics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | - Kumaravelu Jagavelu
- Pharmacology Division, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, India
| | - Madhu Dikshit
- Pharmacology Division, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, India
- Non-Communicable Diseases Division, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|