1
|
Wu X, Zhang Z, Li J, Zong J, Yuan L, Shu L, Cheong LY, Huang X, Jiang M, Ping Z, Xu A, Hoo RL. Chchd10: A Novel Metabolic Sensor Modulating Adipose Tissue Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408763. [PMID: 39985288 PMCID: PMC12005791 DOI: 10.1002/advs.202408763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/30/2024] [Indexed: 02/24/2025]
Abstract
Dysregulation of adipose tissue (AT) homeostasis in obesity contributes to metabolic stress and disorders. Here, we identified that Coiled-coil-helix-coiled-coil-helix domain containing 10 (Chchd10) is a novel regulator of AT remodeling upon excess energy intake. Chchd10 is significantly reduced in the white adipose tissue (WAT) of mice in response to high-fat diet (HFD) feeding. AT-Chchd10 deficiency accelerates adipogenesis predominantly in subcutaneous AT of mice to store excess energy in response to short-term HFD feeding while upregulates glutathione S-transferase A4 (GSTA4) to facilitate 4-HNE clearance mainly in visceral AT to prevent protein carbonylation-induced cell dysfunction after long-term HFD feeding. Hence, Chchd10 deficiency attenuates diet-induced obesity and related metabolic disorders in mice. Mechanistically, Chchd10 deficiency enhances adipogenesis and GSTA4 expression by activating TDP43/Raptor/p62/Keap1/NRF2 axis. Notably, the beneficial effect of Chchd10 deficiency is eliminated in hypertrophic adipocytes, where p62 is strikingly reduced. Collectively, Chchd10 is a metabolic sensor maintaining AT homeostasis, and the loss of p62 in adipose tissue under obese conditions impairs Chchd10-mediated AT remodeling.
Collapse
Affiliation(s)
- Xiaoping Wu
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
- Department of Pharmacology and PharmacyThe University of Hong KongHong Kong SARChina
| | - Zixuan Zhang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
- Department of Pharmacology and PharmacyThe University of Hong KongHong Kong SARChina
| | - Jingjing Li
- Department of Rehabilitation SciencesFaculty of Health and Social SciencesHong Kong Polytechnic UniversityHong Kong SARChina
| | - Jiuyu Zong
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
- Department of Pharmacology and PharmacyThe University of Hong KongHong Kong SARChina
| | - Lufengzi Yuan
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
- Department of Pharmacology and PharmacyThe University of Hong KongHong Kong SARChina
| | - Lingling Shu
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerDepartment of Hematological OncologySun Yat‐sen University Cancer CenterChina
- Department of MedicineThe University of Hong KongHong Kong SARChina
| | - Lai Yee Cheong
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
- Department of MedicineThe University of Hong KongHong Kong SARChina
| | - Xiaowen Huang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
- Department of Pharmacology and PharmacyThe University of Hong KongHong Kong SARChina
| | - Mengxue Jiang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
- Department of Pharmacology and PharmacyThe University of Hong KongHong Kong SARChina
| | - Zhihui Ping
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
- Department of Pharmacology and PharmacyThe University of Hong KongHong Kong SARChina
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
- Department of MedicineThe University of Hong KongHong Kong SARChina
| | - Ruby L.C. Hoo
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
- Department of Pharmacology and PharmacyThe University of Hong KongHong Kong SARChina
| |
Collapse
|
2
|
Singh G, Viramgami A, Makwana B, Mishra S, Dave V, Soundararajan S. Obesity, oxidative stress, and the moderating role of overcommitment: An exploratory analysis in female nurses. Work 2025:10519815251324000. [PMID: 40101259 DOI: 10.1177/10519815251324000] [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: 03/20/2025] Open
Abstract
BackgroundObesity and job stress are established contributors to oxidative stress, which is associated with various adverse health outcomes. However, the moderating role of job stress in the relationship between obesity and oxidative stress remains inadequately understood.ObjectivesThis study explored whether job stress, specifically the extrinsic and intrinsic components of the Effort Reward Imbalance (ERI) model, moderates the relationship between obesity and oxidative stress among female nurses.MethodsWe conducted an exploratory analysis from a cross-sectional study involving female nurses at a tertiary hospital in Western India. The study assessed the prevalence of obesity and overweight based on the suggested criteria for the Asian Indian population. We examined correlations among obesity markers and oxidative stress markers and explored if there is a moderator role of job stress on the relationship between obesity and oxidative stress.ResultsThe combined prevalence of overweight and obesity in the study population was 74%. Significant positive correlations were found between age, obesity-related parameters (Body-mass Index, Percentage Body Fat, Waist-Hip Ratio), and oxidative stress markers (Protein carbonyl content and Glutathione S-Transferase). The relationship between obesity and oxidative stress markers was moderated by overcommitment. Nurses with marked overcommitment exhibited stronger associations between age, obesity, and oxidative stress markers compared to those without marked overcommitment.ConclusionsOur findings suggest that overcommitment moderates the relationship between obesity and oxidative stress, highlighting the need to address intrinsic as well as extrinsic work-related factors in interventions targeting obesity among nurses. Future studies should confirm these findings and explore mechanisms to develop targeted interventions.
Collapse
Affiliation(s)
- Gyanendra Singh
- Toxicology Laboratory, Biological Sciences, ICMR-National Institute of Occupational Health, Ahmedabad, India
| | - Ankit Viramgami
- Clinical Epidemiology, Health Sciences, ICMR-National Institute of Occupational Health, Ahmedabad, India
| | - Bela Makwana
- Clinical Epidemiology, Health Sciences, ICMR-National Institute of Occupational Health, Ahmedabad, India
| | - Sukhdev Mishra
- Clinical Epidemiology, Health Sciences, ICMR-National Institute of Occupational Health, Ahmedabad, India
| | - Viral Dave
- Department of Community Medicine, GCS Medical College, Ahmedabad, India
| | - Soundarya Soundararajan
- Clinical Epidemiology, Health Sciences, ICMR-National Institute of Occupational Health, Ahmedabad, India
| |
Collapse
|
3
|
Liang IC, Gilardoni E, Berdaweel IA, Carter KD, Anderson EJ. Low Plasma Carnosinase-1 Activity in Patients with Left Ventricular Systolic Dysfunction: Implications for Carnosine Therapy in Heart Failure. Int J Mol Sci 2025; 26:2608. [PMID: 40141250 PMCID: PMC11942450 DOI: 10.3390/ijms26062608] [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: 12/31/2024] [Revised: 03/01/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Therapeutic efficacy of histidyl dipeptides such as carnosine is hampered by circulating carnosinase-1 (CN1), which catalyzes carnosine's hydrolysis and degradation. Prior reports suggest that oral carnosine may improve cardiometabolic parameters in patients with heart failure (HF), but whether CN1 activity is affected by HF is unknown. Here, we measured CN1 content and carnosine degradation rate (CDR) in preoperative plasma samples from a cohort of patients (n = 138) undergoing elective cardiac surgery to determine whether plasma CN1 and/or CDR varied with left ventricular (LV) systolic dysfunction. CN1 content was normally distributed in the cohort, but plasma CDR displayed a quasi-bimodal distribution into high- (>2 nmol/(h*μL)) and low-activity (≤2 nmol/(h*μL)) clusters. Multivariable analysis confirmed female sex, diabetes and LV systolic dysfunction was associated with the low-activity CDR cluster. Although CN1 content did not differ, logistic regression analysis revealed that CDR and CN1-specific activity (CDR/CN1 content) was significantly lower in patients with both moderate (ejection fraction, EF ≥ 35 to <50%) and severe LV systolic dysfunction (EF < 35%) compared with patients in the normal range (EF ≥ 50%). These findings suggest that plasma CN1 activity is regulated by factors independent of expression, and that a decline in LV systolic function is associated with low CN1 activity. Further studies are needed to delineate specific mechanisms controlling CN1 expression and activity, which will facilitate the development of carnosine and other histidyl dipeptide therapies for cardiometabolic disorders such as HF.
Collapse
Affiliation(s)
- I-Chau Liang
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA; (I.-C.L.); (I.A.B.)
| | - Ettore Gilardoni
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA; (I.-C.L.); (I.A.B.)
| | - Islam A. Berdaweel
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA; (I.-C.L.); (I.A.B.)
- Department of Clinical Pharmacy and Pharmacy Practice, Yarmouk University, Irbid 21163, Jordan
| | - Knute D. Carter
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, IA 52242, USA;
| | - Ethan J. Anderson
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA; (I.-C.L.); (I.A.B.)
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
4
|
Merali C, Quinn C, Huffman KM, Pieper CF, Bogan JS, Barrero CA, Merali S. Sustained caloric restriction potentiates insulin action by activating prostacyclin synthase. Obesity (Silver Spring) 2024; 32:2286-2298. [PMID: 39420421 PMCID: PMC12034231 DOI: 10.1002/oby.24150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE Caloric restriction (CR) is known to enhance insulin sensitivity and reduce the risk of metabolic disorders; however, its molecular mechanisms are not fully understood. This study aims to elucidate specific proteins and pathways responsible for these benefits. METHODS We examined adipose tissue from participants in the Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy Phase 2 (CALERIE 2) study, comparing proteomic profiles from individuals after 12 and 24 months of CR with baseline and an ad libitum group. Biochemical and cell-specific physiological approaches complemented these analyses. RESULTS Our data revealed that CR upregulates prostacyclin synthase (PTGIS) in adipose tissue, an enzyme crucial for producing prostacyclin (PGI2). PGI2 improves the ability of insulin to stimulate the tether-containing UBX domain for GLUT4 (TUG) cleavage pathway, which is essential for glucose uptake regulation. Additionally, iloprost, a PGI2 analog, was shown to increase insulin receptor density on cell membranes, increasing glucose uptake in human adipocytes. CR also reduces carbonylation of GLUT4, a modification that is detrimental to GLUT4 function. CONCLUSIONS CR enhances insulin sensitivity by promoting PTGIS expression and stimulating the TUG cleavage pathway, leading to increased GLUT4 translocation to the cell surface and decreased GLUT4 carbonylation. These findings shed light on the complex molecular mechanisms through which CR favorably impacts insulin sensitivity and metabolic health.
Collapse
Affiliation(s)
- Carmen Merali
- Temple University School of Pharmacy, Philadelphia, Pennsylvania, USA
| | - Connor Quinn
- Temple University School of Pharmacy, Philadelphia, Pennsylvania, USA
| | - Kim M. Huffman
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Carl F. Pieper
- Duke Center for Aging and Human Development, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jonathan S. Bogan
- Section of Endocrinology and Metabolism, Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Carlos A. Barrero
- Temple University School of Pharmacy, Philadelphia, Pennsylvania, USA
| | - Salim Merali
- Temple University School of Pharmacy, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Cota-Magaña AI, Vazquez-Moreno M, Rocha-Aguado A, Ángeles-Mejía S, Valladares-Salgado A, Díaz-Flores M, López-Díazguerrero NE, Cruz M. Obesity Is Associated with Oxidative Stress Markers and Antioxidant Enzyme Activity in Mexican Children. Antioxidants (Basel) 2024; 13:457. [PMID: 38671905 PMCID: PMC11047352 DOI: 10.3390/antiox13040457] [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] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The relationship between metabolic disorders and oxidative stress is still controversial in the child population. The present cross-sectional study aimed to analyze the associations between obesity, cardiometabolic traits, serum level of carbonylated proteins (CPs), malondialdehyde (MDA), and the enzyme activity of catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx) in children from Mexico City (normal weight: 120; obesity: 81). Obesity resulted in being positively associated with CAT (β = 0.05 ± 0.01, p = 5.0 × 10-3) and GPx (β = 0.13 ± 0.01, p = 3.7 × 10-19) enzyme activity. A significant interaction between obesity and sex was observed in MDA and SOD enzymatic activity (PMDA = 0.03; PSOD = 0.04). The associations between obesity, MDA level, and SOD enzyme activity were only significant in boys (boys: PMDA = 3.0 × 10-3; PSOD = 7.0 × 10-3; girls: p ≥ 0.79). In both children with normal weight and those with obesity, CP levels were positively associated with SOD enzyme activity (PNormal-weight = 2.2 × 10-3; PObesity = 0.03). In conclusion, in Mexican children, obesity is positively associated with CAT and GPx enzyme activity, and its associations with MDA levels and SOD enzyme activity are sex-specific. Therefore, CP level is positively related to SOD enzyme activity independently of body weight.
Collapse
Affiliation(s)
- Ana Isabel Cota-Magaña
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (A.I.C.-M.); (M.V.-M.)
- Programa de Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Mexico City 04960, Mexico
- Laboratorio de Bioenergética y Envejecimiento Celular, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana Unidad Iztapalapa, Mexico City 09340, Mexico
| | - Miguel Vazquez-Moreno
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (A.I.C.-M.); (M.V.-M.)
| | - Andrés Rocha-Aguado
- OOAD Ciudad de México Norte, Unidad de Medicina Familiar No. 23, Instituto Mexicano del Seguro Social, Mexico City 07070, Mexico
| | - Selene Ángeles-Mejía
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (A.I.C.-M.); (M.V.-M.)
| | - Adán Valladares-Salgado
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (A.I.C.-M.); (M.V.-M.)
| | - Margarita Díaz-Flores
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (A.I.C.-M.); (M.V.-M.)
| | - Norma Edith López-Díazguerrero
- Laboratorio de Bioenergética y Envejecimiento Celular, Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana Unidad Iztapalapa, Mexico City 09340, Mexico
| | - Miguel Cruz
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI del Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (A.I.C.-M.); (M.V.-M.)
| |
Collapse
|
6
|
Tuell D, Ford G, Los E, Stone W. The Role of Glutathione and Its Precursors in Type 2 Diabetes. Antioxidants (Basel) 2024; 13:184. [PMID: 38397782 PMCID: PMC10885928 DOI: 10.3390/antiox13020184] [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: 12/13/2023] [Revised: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Type 2 diabetes (T2D) is a major worldwide health crisis affecting about 6.2% of the world's population. Alarmingly, about one in five children in the USA have prediabetes. Glutathione (GSH) and its precursors play a promising role in the prevention and management of type T2D. Oxidative stress (OxS) is a probable factor in both T2D initiation and progression. GSH is the major cytosolic water-soluble chemical antioxidant and emerging evidence supports its role in improving T2D outcomes. Dietary supplementation with N-acetyl-cysteine (NAC) and/or glycine (GLY), which are GSH precursors, has also been studied for possible beneficial effects on T2D. This review will focus on the underlying pathophysiological and molecular mechanisms linking GSH and its precursors with T2D and OxS. In addition to their traditional antioxidant roles, the in vivo effects of GSH/NAC/GLY supplements will be evaluated for their potential abilities to modulate the complex pro-oxidant pathophysiological factors (e.g., hyperglycemia) driving T2D progression. Positive feedback loops that amplify OxS over long time intervals are likely to result in irreversible T2D micro- and macro-vascular damage. Most clinical studies with GSH/NAC/GLY have focused on adults or the elderly. Future research with pediatric populations should be a high priority since early intervention is critical.
Collapse
|
7
|
Riggs PK, Anderson AM, Tang B, Rubin LH, Morgello S, Marra CM, Gelman BB, Clifford DB, Franklin D, Heaton RK, Ellis RJ, Fennema-Notestine C, Letendre SL. Elevated Plasma Protein Carbonyl Concentration Is Associated with More Abnormal White Matter in People with HIV. Viruses 2023; 15:2410. [PMID: 38140650 PMCID: PMC10747698 DOI: 10.3390/v15122410] [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: 08/17/2023] [Revised: 11/23/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Structural brain abnormalities, including those in white matter (WM), remain common in people with HIV (PWH). Their pathogenesis is uncertain and may reflect multiple etiologies. Oxidative stress is associated with inflammation, HIV, and its comorbidities. The post-translational carbonylation of proteins results from oxidative stress, and circulating protein carbonyls may reflect this. In this cross-sectional analysis, we evaluated the associations between protein carbonyls and a panel of soluble biomarkers of neuronal injury and inflammation in plasma (N = 45) and cerebrospinal fluid (CSF, n = 32) with structural brain MRI. The volume of abnormal WM was normalized for the total WM volume (nAWM). In this multisite project, all regression models were adjusted for the scanner. The candidate covariates included demographics, HIV disease characteristics, and comorbidities. Participants were PWH on virally suppressive antiretroviral therapy (ART) and were mostly white (64.4%) men (88.9%), with a mean age of 56.8 years. In unadjusted analyses, more nAWM was associated with higher plasma protein carbonyls (p = 0.002) and higher CCL2 (p = 0.045). In the adjusted regression models for nAWM, the association with plasma protein carbonyls remained significant (FDR p = 0.018). Protein carbonyls in plasma may be a valuable biomarker of oxidative stress and its associated adverse health effects, including within the central nervous system. If confirmed, these findings would support the hypothesis that reducing oxidative stress could treat or prevent WM injury in PWH.
Collapse
Affiliation(s)
- Patricia K. Riggs
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
| | - Albert M. Anderson
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Bin Tang
- Department of Psychiatry, University of California San Diego, San Diego, CA 92093, USA
| | - Leah H. Rubin
- Departments of Neurology, Psychiatry and Behavioral Sciences, and Epidemiology, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Susan Morgello
- Departments of Neurology, Neuroscience, and Pathology, Mt Sinai School of Medicine, New York, NY 10029, USA
| | - Christina M. Marra
- Department of Neurology, University of Washington, Seattle, WA 98195, USA
| | - Benjamin B. Gelman
- Departments of Pathology, and Neuroscience & Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - David B. Clifford
- Department of Neurology, Washington University in St Louis, St Louis, MO 63110, USA
| | - Donald Franklin
- Department of Psychiatry, University of California San Diego, San Diego, CA 92093, USA
| | - Robert K. Heaton
- Department of Psychiatry, University of California San Diego, San Diego, CA 92093, USA
| | - Ronald J. Ellis
- Department of Psychiatry, University of California San Diego, San Diego, CA 92093, USA
- Department of Neurosciences, University of California San Diego, San Diego, CA 92093, USA
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California San Diego, San Diego, CA 92093, USA
- Department of Radiology, University of California San Diego, San Diego, CA 92093, USA
| | - Scott L. Letendre
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, San Diego, CA 92093, USA
- Department of Psychiatry, University of California San Diego, San Diego, CA 92093, USA
| |
Collapse
|
8
|
Murdolo G, Bartolini D, Tortoioli C, Vermigli C, Piroddi M, Galli F. Accumulation of 4-Hydroxynonenal Characterizes Diabetic Fat and Modulates Adipogenic Differentiation of Adipose Precursor Cells. Int J Mol Sci 2023; 24:16645. [PMID: 38068967 PMCID: PMC10705911 DOI: 10.3390/ijms242316645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Redox imbalance in fat tissue appears to be causative of impaired glucose homeostasis. This "proof-of-concept" study investigated whether the peroxidation by-product of polyunsaturated n-6 fatty acids, namely 4-hydroxynonenal (4-HNE), is formed by, and accumulates in, the adipose tissue (AT) of obese patients with type 2 diabetes (OBT2D) as compared with lean, nondiabetic control subjects (CTRL). Moreover, we studied the effects of 4-HNE on the cell viability and adipogenic differentiation of adipose-derived stem cells (ASCs). Protein-HNE adducts in subcutaneous abdominal AT (SCAAT) biopsies from seven OBT2D and seven CTRL subjects were assessed using Western blot. The effects of 4-HNE were then studied in primary cultures of ASCs, focusing on cell viability, adipogenic differentiation, and the "canonical" Wnt and MAPK signaling pathways. When compared with the controls, the OBT2D patients displayed increased HNE-protein adducts in the SCAAT. The exposure of ASCs to 4-HNE fostered ROS production and led to a time- and concentration-dependent decrease in cell viability. Notably, at concentrations that did not affect cell viability (1 μM), 4-HNE hampered adipogenic ASCs' differentiation through a timely-regulated activation of the Wnt/β-catenin, p38MAPK, ERK1/2- and JNK-mediated pathways. These "hypothesis-generating" data suggest that the increased accumulation of 4-HNE in the SCAAT of obese patients with type 2 diabetes may detrimentally affect adipose precursor cell differentiation, possibly contributing to the obesity-associated derangement of glucose homeostasis.
Collapse
Affiliation(s)
- Giuseppe Murdolo
- Department of Internal Medicine, Endocrinology and Metabolism, Azienda Ospedaliera S. Maria Misericordia, University of Perugia, Piazzale Gambuli, I-06081 Perugia, Italy (C.V.)
| | - Desirée Bartolini
- Department of Pharmaceutical Sciences, Section of Applied Biochemistry and Nutritional Sciences, University of Perugia, I-06081 Perugia, Italy (F.G.)
| | - Cristina Tortoioli
- Department of Internal Medicine, Endocrinology and Metabolism, Azienda Ospedaliera S. Maria Misericordia, University of Perugia, Piazzale Gambuli, I-06081 Perugia, Italy (C.V.)
| | - Cristiana Vermigli
- Department of Internal Medicine, Endocrinology and Metabolism, Azienda Ospedaliera S. Maria Misericordia, University of Perugia, Piazzale Gambuli, I-06081 Perugia, Italy (C.V.)
| | | | - Francesco Galli
- Department of Pharmaceutical Sciences, Section of Applied Biochemistry and Nutritional Sciences, University of Perugia, I-06081 Perugia, Italy (F.G.)
| |
Collapse
|
9
|
Zhu X, Ren T, Xiong Q, Lin Z, Lin X, Lin G. Salidroside alleviates diet-induced obesity and insulin resistance by activating Nrf2/ARE pathway and enhancing the thermogenesis of adipose tissues. Food Sci Nutr 2023; 11:4735-4744. [PMID: 37576042 PMCID: PMC10420790 DOI: 10.1002/fsn3.3450] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 08/15/2023] Open
Abstract
Recent reports suggest that salidroside protects cardiomyocytes from oxidative injury and stimulates glucose uptake by skeletal muscle cells. Despite these findings, the therapeutic potential of salidroside in the treatment of obesity and insulin resistance remains uncertain and requires further investigation. In the present study, the treatment effect of salidroside on the onset and development of the obese phenotype and insulin resistance as well as the underlying mechanisms was investigated using long-term high-fat diet-induced obese mice supplemented with salidroside. We used biochemical kits to determine serum biochemical parameters (including triacylglycerol, total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, fasting glucose, and insulin). The results show that salidroside-supplemented animals showed better glucose tolerance and insulin sensitivity, decreased blood lipids, and weight gain (p < .05). Protein expression of p-Nrf2 and Nrf2 was analyzed by western blotting, and the mRNA levels of thermogenic-related genes (Ucp1, Pgc1a, Prdm16, and Cidea) were detected by quantitative RT-PCR. The results show an improvement in lipid peroxidation and Nrf2/ARE signaling, as well as an increased expression of the Ucp1, Pgc1a, Prdm16, and Cidea (p < .05). Our evidence suggests that salidroside alleviates diet-induced obesity and insulin resistance potentially by activating Nrf2/ARE pathway and enhancing the thermogenesis of adipose tissues. This induction represents a potential technique for the management of comorbidities related to obesity and its prevention.
Collapse
Affiliation(s)
- Xiaozhen Zhu
- Department of PharmacyThe Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Ting Ren
- College of Life and Environmental ScienceWenzhou UniversityWenzhouChina
| | - Qiushuang Xiong
- Department of PharmacyThe Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Zhengfeng Lin
- Department of PharmacyThe Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xiaoxiao Lin
- Department of PharmacyThe Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Guangyong Lin
- Department of PharmacyThe Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
10
|
Nelson TJ, Xu Y. Sting and p53 DNA repair pathways are compromised in Alzheimer's disease. Sci Rep 2023; 13:8304. [PMID: 37221295 PMCID: PMC10206146 DOI: 10.1038/s41598-023-35533-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/19/2023] [Indexed: 05/25/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. A common finding in AD is DNA damage. Double-strand DNA breaks (DSBs) are particularly hazardous to neurons because their post-mitotic state forces neurons to rely on error-prone and potentially mutagenic mechanisms to repair DNA breaks. However, it remains unclear whether DNA damage results from increased DNA damage or failure of DNA repair. Oligomerization of the tumor suppressor protein p53 is an essential part of DSB repair, and p53 phosphorylated on S15 is an indicator of DNA damage. We report that the monomer:dimer ratio of phosphorylated (S15) p53 is increased by 2.86-fold in temporal lobes of AD patients compared to age-matched controls, indicating that p53 oligomerization is compromised in AD. In vitro oxidation of p53 with 100 nM H2O2 produced a similar shift in the monomer:dimer ratio. A COMET test showed a higher level of DNA degradation in AD consistent with double-strand DNA damage or inhibition of repair. Protein carbonylation was also elevated (190% of control), indicating elevated oxidative stress in AD patients. Levels of the DNA repair support protein 14-3-3σ, γ-H2AX, a phosphorylated histone marking double strand DNA breaks, and phosphorylated ataxia telangiectasia mutated (ATM) protein were all increased. cGAS-STING-interferon signaling was impaired in AD and was accompanied by a depletion of STING protein from Golgi and a failure to elevate interferon despite the presence of DSBs. The results suggest that oxidation of p53 by ROS could inhibit the DDR and decrease its ability to orchestrate DSB repair by altering the oligomerization state of p53. The failure of immune-stimulated DNA repair may contribute to cell loss in AD and suggests new therapeutic targets for AD.
Collapse
Affiliation(s)
- Thomas J Nelson
- Department of Neurology, Marshall University Joan C. Edwards School of Medicine, Huntington, WV, 25704, USA.
| | - Yunhui Xu
- Department of Neurology, Marshall University Joan C. Edwards School of Medicine, Huntington, WV, 25704, USA
| |
Collapse
|
11
|
Li H, Beg OU, Rafie AR, Kanwal S, Ovalle-Cisneros A, Faison MO, Siddiqui RA. Characterization of Green and Yellow Papaya ( Carica papaya) for Anti-Diabetic Activity in Liver and Myoblast Cells and Wound-Healing Activity in Fibroblast Cells. Nutrients 2023; 15:1929. [PMID: 37111148 PMCID: PMC10142885 DOI: 10.3390/nu15081929] [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: 03/20/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Obesity and diabetes, often characterized as "metabolic syndrome", have been recognized as two of the most important public health issues worldwide. The objective of the present research was to evaluate green and yellow papaya for anti-oxidation and anti-diabetic properties. Leaves, skin, pulp, and seed samples from papayas were freeze-dried and then extracted in water or 80% methanol. The extracts were used to determine total polyphenolic content and anti-oxidation activities, and to determine biological activities, including glucose uptake, Glut-2 expression, triglyceride reduction, and wound-healing activity. Our data demonstrated that methanol and water extracts of green and yellow papaya have similar concentrations of polyphenols in skin (10-20 mg/g dry powder), leaf (25-30 mg/g dry powder), and pulp (1-3 mg/g dry powder) fractions. However, both methanol and water extracts of seeds from yellow papaya have substantially higher concentrations of polyphenols compared to green papaya. Both water and methanol extracts of yellow papaya exhibited higher anti-oxidation activity compared to green papaya in skin (50-60%), pulp (200-300%), and seeds (10-800%). Old leaves also showed greater anti-oxidation activity (30-40%) compared to new leaves. Pulp extracts from both yellow and green papaya stimulated greater glucose uptake, but only pulp from green papaya stimulated glucose uptake in muscle cells. Similarly, pulp extract stimulated glucose transporter Glut-2 expression in liver cells. The skin, pulp, and seeds of green or yellow papaya showed triglyceride-lowering activity in liver cells by 60-80%, but samples taken from yellow papaya had a more potent effect. Seeds from both green and yellow papaya significantly stimulated the migration of fibroblasts in the wounded area by 2-2.5-fold compared to the untreated control. Consistent with these data, seeds from both green and yellow papaya also significantly stimulated collagen synthesis in fibroblast cells by almost 3-fold. In conclusion, our data indicate that different parts of papaya produce stimulatory effects on glucose uptake, Glut-2 expression, TG reduction, and wound-healing activities. This study concludes that different parts of the papaya can be beneficial for preventing diabetes and diabetes-related wound healing.
Collapse
Affiliation(s)
- Haiwen Li
- Food Chemistry and Nutrition Science Laboratory, Agricultural Research Station, College of Agriculture, Virginia State University, Petersburg, VA 23806, USA; (H.L.); (O.U.B.); (S.K.)
| | - Obaid Ullah Beg
- Food Chemistry and Nutrition Science Laboratory, Agricultural Research Station, College of Agriculture, Virginia State University, Petersburg, VA 23806, USA; (H.L.); (O.U.B.); (S.K.)
| | - Ahmed Reza Rafie
- Cooperate Extension, College of Agriculture, Virginia State University, Petersburg, VA 23806, USA;
| | - Sadia Kanwal
- Food Chemistry and Nutrition Science Laboratory, Agricultural Research Station, College of Agriculture, Virginia State University, Petersburg, VA 23806, USA; (H.L.); (O.U.B.); (S.K.)
| | - Alexandra Ovalle-Cisneros
- Department of Biology, College of Natural Sciences, Virginia State University, Petersburg, VA 23806, USA; (A.O.-C.); (M.O.F.)
| | - Milton Omar Faison
- Department of Biology, College of Natural Sciences, Virginia State University, Petersburg, VA 23806, USA; (A.O.-C.); (M.O.F.)
| | - Rafat Ali Siddiqui
- Food Chemistry and Nutrition Science Laboratory, Agricultural Research Station, College of Agriculture, Virginia State University, Petersburg, VA 23806, USA; (H.L.); (O.U.B.); (S.K.)
| |
Collapse
|
12
|
Kitamura Y, Oikawa S, Chang J, Mori Y, Ichihara G, Ichihara S. Carbonylated Proteins as Key Regulators in the Progression of Metabolic Syndrome. Antioxidants (Basel) 2023; 12:antiox12040844. [PMID: 37107219 PMCID: PMC10135001 DOI: 10.3390/antiox12040844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Based on the known role of oxidative stress in the pathogenesis and progression of metabolic syndrome, we used two-dimensional gel electrophoresis with immunochemical detection of protein carbonyls (2D-Oxyblot) to characterize the carbonylated proteins induced by oxidative stress in spontaneously hypertensive rats/NDmcr-cp (CP), an animal model of metabolic syndrome. We also profiled the proteins that showed change of expression levels in their epididymal adipose tissue at the pre-symptomatic (6-week-old) and the symptomatic (25-week-old) stages of the metabolic syndrome. Two-dimensional fluorescence difference gel electrophoresis (2D-DIGE) combined with matrix-assisted laser desorption ionization time-of-flight tandem mass spectrometry (MALDI-TOF/TOF MS) was used to analyze proteins extracted from the epididymal adipose tissue. The up-regulated proteins identified at the pre-symptomatic stage were mainly associated with ATP production and redox reaction, while the down-regulated proteins found at the symptomatic stage were involved in antioxidant activity and the tricarboxylic acid (TCA) cycle. Further analysis using the 2D-Oxyblot showed significantly high carbonylation levels of gelsolin and glycerol-3-phosphate dehydrogenase [NAD+] at the symptomatic stage. These results suggest that reduced antioxidant capacity underlies the increased oxidative stress state in the metabolic syndrome. The identified carbonylated proteins, including gelsolin, are potential targets that may act as key regulators in the progression of the metabolic syndrome.
Collapse
Affiliation(s)
- Yuki Kitamura
- Department of Molecular and Environmental Medicine, Mie University Graduate School of Medicine, Tsu 514-8507, Japan
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke 329-0498, Japan
| | - Shinji Oikawa
- Department of Molecular and Environmental Medicine, Mie University Graduate School of Medicine, Tsu 514-8507, Japan
| | - Jie Chang
- Graduate School of Regional Innovation Studies, Mie University, Tsu 514-8507, Japan
| | - Yurie Mori
- Department of Molecular and Environmental Medicine, Mie University Graduate School of Medicine, Tsu 514-8507, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Tokyo University of Sciences, Noda 278-8510, Japan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke 329-0498, Japan
- Graduate School of Regional Innovation Studies, Mie University, Tsu 514-8507, Japan
| |
Collapse
|
13
|
Rasool A, Mahmoud T, O’Tierney-Ginn P. Lipid Aldehydes 4-Hydroxynonenal and 4-Hydroxyhexenal Exposure Differentially Impact Lipogenic Pathways in Human Placenta. BIOLOGY 2023; 12:527. [PMID: 37106728 PMCID: PMC10135722 DOI: 10.3390/biology12040527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/16/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023]
Abstract
Long chain polyunsaturated fatty acids (LCPUFAs), such as the omega-6 (n-6) arachidonic acid (AA) and n-3 docosahexanoic acid (DHA), have a vital role in normal fetal development and placental function. Optimal supply of these LCPUFAs to the fetus is critical for improving birth outcomes and preventing programming of metabolic diseases in later life. Although not explicitly required/recommended, many pregnant women take n-3 LCPUFA supplements. Oxidative stress can cause these LCPUFAs to undergo lipid peroxidation, creating toxic compounds called lipid aldehydes. These by-products can lead to an inflammatory state and negatively impact tissue function, though little is known about their effects on the placenta. Placental exposure to two major lipid aldehydes, 4-hydroxynonenal (4-HNE) and 4-hydroxyhexenal (4-HHE), caused by peroxidation of the AA and DHA, respectively, was examined in the context of lipid metabolism. We assessed the impact of exposure to 25 μM, 50 μM and 100 μM of 4-HNE or 4-HHE on 40 lipid metabolism genes in full-term human placenta. 4-HNE increased gene expression associated with lipogenesis and lipid uptake (ACC, FASN, ACAT1, FATP4), and 4-HHE decreased gene expression associated with lipogenesis and lipid uptake (SREBP1, SREBP2, LDLR, SCD1, MFSD2a). These results demonstrate that these lipid aldehydes differentially affect expression of placental FA metabolism genes in the human placenta and may have implications for the impact of LCPUFA supplementation in environments of oxidative stress.
Collapse
|
14
|
Basu T, Selman A, Reddy AP, Reddy PH. Current Status of Obesity: Protective Role of Catechins. Antioxidants (Basel) 2023; 12:antiox12020474. [PMID: 36830032 PMCID: PMC9952428 DOI: 10.3390/antiox12020474] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Obesity is a growing health concern in today's society. Current estimates indicate that obesity occurs in both adults and young people. Recent research also found that the Hispanic population in the U.S. is 1.9 times more likely to be overweight as compared to their non-Hispanic population. Obesity is a multifactorial disease that has a variety of causes. All current treatment options incorporate dietary changes aimed at establishing a negative energy balance. According to current scientific research, multiple factors are involved with the development of obesity, including genetic, biochemical, psychological, environmental, behavioral, and socio-demographic factors. The people who suffer from obesity are far more likely to suffer serious health problems, such as stroke, diabetes, lung disease, bone and joint disease, cancer, heart disease, neurological disorders, and poor mental health. Studies indicate that multiple cellular changes are implicated in the progression of obesity, mitochondrial dysfunction, deregulated microRNAs, inflammatory changes, hormonal deregulation, and others. This article highlights the role that oxidative stress plays in obesity and current obesity-prevention techniques with an emphasis on the impact of catechins to prevent and treat obesity.
Collapse
Affiliation(s)
- Tanisha Basu
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Ashley Selman
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Arubala P. Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Correspondence: ; Tel.: +1-806-743-3194; Fax: +1-806-743-2334
| |
Collapse
|
15
|
Kaur S, Rubal, Kaur S, Kaur A, Kaur S, Gupta S, Mittal S, Dhiman M. A cross-sectional study to correlate antioxidant enzymes, oxidative stress and inflammation with prevalence of hypertension. Life Sci 2023; 313:121134. [PMID: 36544300 DOI: 10.1016/j.lfs.2022.121134] [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: 01/17/2022] [Revised: 09/19/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
Abstract
AIMS Hypertension a multifactorial consequence of environmental factors, life style and genetics is the well-recognized risk factor contributing to coronary heart diseases. The antioxidant imbalance, excessive reactive oxygen species (ROS) leads to oxidative stress which is pivotal in progression of hypertension. The present study aims to understand the complex interaction between oxidative stress, inflammation and antioxidant system which is crucial to maintain cellular homeostasis which further can exaggerate hypertension pathophysiology. MATERIALS AND METHODS The metabolic profile of hypertensive and normotensive subjects from Malwa region, Punjab was compared by estimating lipid profile, cardiac, hepatic and renal markers. The oxidative stress markers (protein carbonyls and lipid peroxidation), inflammatory markers (Nitric oxide, Myeloperoxidase and advanced oxygen protein products), and antioxidant enzymes (Superoxide Dismutase, Catalase, and Total Antioxidant Capacity) were analyzed. KEY FINDINGS It is observed that the metabolic markers are altered in hypertensive subjects which further these subjects showed increased oxidative, inflammatory profile and compromised antioxidant status when compared with normotensive subjects. Co-relation analysis validated the involvement of inflammation and oxidative stress in impaired endothelial function and vital organ damage. SIGNIFICANCE OF STUDY These markers may act as early indicators of hypertension which usually do not show any physical symptoms, thus can be diagnosed and treated at the earliest. The current study suggests that disturbed homeostasis, a consequence of altered interaction between antioxidant system and inflammatory events raises the oxidative stress levels which eventually leads to hypertension and associated complications. These indicators can serve as early indicators of future chronic complications of hypertension.
Collapse
Affiliation(s)
- Sukhchain Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab Bathinda, India
| | - Rubal
- Department of Microbiology, School of Basic Sciences, Central University of Punjab Bathinda, India
| | - Satveer Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab Bathinda, India
| | - Amandeep Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab Bathinda, India
| | - Sandeep Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab Bathinda, India
| | - Sushil Gupta
- Department of Microbiology, School of Basic Sciences, Central University of Punjab Bathinda, India
| | - Sunil Mittal
- Department of Environmental Science and Technology, School of Environment and Earth Sciences, Central University of Punjab Bathinda, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic Sciences, Central University of Punjab Bathinda, India.
| |
Collapse
|
16
|
Liu J, He L, Wang A, Lv Y, He H, Wang C, Xiong K, Zhao L. Oxidative balance score reflects vascular endothelial function of Chinese community dwellers. Front Physiol 2023; 14:1076327. [PMID: 37138670 PMCID: PMC10150015 DOI: 10.3389/fphys.2023.1076327] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
Background: The oxidative balance score (OBS) is a composite estimate of the overall pro- and antioxidant risk status in an individual. The aim of this study is to explore the association between the OBS and vascular endothelial function in Chinese community dwellers. Methods: In total, 339 community dwelling adults (aged 20-75 years) were recruited in this study. The overall OBS was calculated on the basis of 16 pro- and antioxidant factors related to diet (measured by fasting blood samples) and lifestyle (evaluated by questionnaires). The dietary OBS and lifestyle OBS were calculated on the basis of the corresponding components. Serum iso-prostaglandin F2α (FIP) was measured to evaluate the oxidative stress degree, and brachial artery blood flow-mediated dilation (FMD) was measured for vascular endothelial function. The FIP and FMD levels were dichotomized as "low" or "high" using the corresponding median values (low FIP, n = 159; high FIP, n = 180; low FMD, n = 192; and high FMD, n = 147). The components of the OBS were compared between the stratified FIP and FMD groups. Logistic regression was used to analyze the OBS associations with FIP and FMD. Results: The higher overall OBS and dietary OBS were associated with lower FIP (p < 0.001), whereas the higher overall OBS (p < 0.01) and dietary OBS (p < 0.05) were associated with higher FMD. The lifestyle OBS was not associated with FIP and FMD (p > 0.05). Except for the body mass index (BMI) and low physical activity, all other OBS components were significantly different between the low FIP and high FIP groups (p < 0.05). Four diet-related antioxidants (α-carotene, zeaxanthin, α-tocopherol, and γ-tocopherol) showed significant differences between the high and low FMD groups (p < 0.05). Conclusion: The decreasing OBS level was associated with low endothelial function and high oxidative stress. The dietary OBS, rather than the lifestyle OBS, was more closely associated with endothelial function.
Collapse
Affiliation(s)
- Jianhua Liu
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
- College of Physical Education, WeiFang University, WeiFang, Shandong, China
| | - Lingxiao He
- School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Aozhe Wang
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Yuanyuan Lv
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Hui He
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Chenghao Wang
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Kaiyu Xiong
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Li Zhao
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
- *Correspondence: Li Zhao,
| |
Collapse
|
17
|
Kaur S, Garg N, Rubal R, Dhiman M. Correlative study on heavy metal-induced oxidative stress and hypertension among the rural population of Malwa Region of Punjab, India. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:90948-90963. [PMID: 35881282 DOI: 10.1007/s11356-022-20850-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/11/2022] [Indexed: 06/15/2023]
Abstract
Heavy metal-induced toxicity contributes to the progression of various metabolic disorders and possible mechanisms involved in disease progression are not well established. In this study, the correlation of heavy metal exposure and hypertension have been demonstrated. The results showed that in hypertensive subjects, the lipid profiles (triglycerides, LDL-C, HDL-C, and total cholesterol) and cardiac markers (CK-MB and LDH) were altered abruptly. As a consequence of heavy- induced oxidative stress, the oxidants (TBARS and protein carbonyls) and antioxidants (SOD, GSH, and TAC) were significantly increased and decreased, respectively in hypertension subjects. The concentrations of heavy metals (Pb, Cd, and As) exceeded the permissible limits in hypertensive subjects. The Nrf-2 genotyping indicated that heavy metals may induce mutations at molecular level. The results of correlation analysis revealed that the heavy metals interact with cellular components and interfere with metabolic processes which then results in disturbed lipid profile, enhanced oxidative stress, and reduced antioxidant status. The current study systematically estimated the association of hair and nail heavy metal concentrations with hypertension among the population residing in the Malwa region of Punjab. The proposed study highlighted that heavy metals act as a silent risk factor in the hypertension progression in the population of Malwa region. Future studies are required to confirm current findings and further scrutinize the effect of heavy metals exposure in early adulthood, early, and late mid-life to develop metabolic complications such as hypertension.
Collapse
Affiliation(s)
- Sukhchain Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Neha Garg
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Rubal Rubal
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
18
|
Kleiboeker B, Lodhi IJ. Peroxisomal regulation of energy homeostasis: Effect on obesity and related metabolic disorders. Mol Metab 2022; 65:101577. [PMID: 35988716 PMCID: PMC9442330 DOI: 10.1016/j.molmet.2022.101577] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/01/2022] [Accepted: 08/16/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Peroxisomes are single membrane-bound organelles named for their role in hydrogen peroxide production and catabolism. However, their cellular functions extend well beyond reactive oxygen species (ROS) metabolism and include fatty acid oxidation of unique substrates that cannot be catabolized in mitochondria, and synthesis of ether lipids and bile acids. Metabolic functions of peroxisomes involve crosstalk with other organelles, including mitochondria, endoplasmic reticulum, lipid droplets and lysosomes. Emerging studies suggest that peroxisomes are important regulators of energy homeostasis and that disruption of peroxisomal functions influences the risk for obesity and the associated metabolic disorders, including type 2 diabetes and hepatic steatosis. SCOPE OF REVIEW Here, we focus on the role of peroxisomes in ether lipid synthesis, β-oxidation and ROS metabolism, given that these functions have been most widely studied and have physiologically relevant implications in systemic metabolism and obesity. Efforts are made to mechanistically link these cellular and systemic processes. MAJOR CONCLUSIONS Circulating plasmalogens, a form of ether lipids, have been identified as inversely correlated biomarkers of obesity. Ether lipids influence metabolic homeostasis through multiple mechanisms, including regulation of mitochondrial morphology and respiration affecting brown fat-mediated thermogenesis, and through regulation of adipose tissue development. Peroxisomal β-oxidation also affects metabolic homeostasis through generation of signaling molecules, such as acetyl-CoA and ROS that inhibit hydrolysis of stored lipids, contributing to development of hepatic steatosis. Oxidative stress resulting from increased peroxisomal β-oxidation-generated ROS in the context of obesity mediates β-cell lipotoxicity. A better understanding of the roles peroxisomes play in regulating and responding to obesity and its complications will provide new opportunities for their treatment.
Collapse
Affiliation(s)
- Brian Kleiboeker
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO 63110 USA.
| |
Collapse
|
19
|
Oh JM, Chun S. Ginsenoside CK Inhibits the Early Stage of Adipogenesis via the AMPK, MAPK, and AKT Signaling Pathways. Antioxidants (Basel) 2022; 11:1890. [PMID: 36290613 PMCID: PMC9598147 DOI: 10.3390/antiox11101890] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/15/2022] [Accepted: 09/21/2022] [Indexed: 08/25/2023] Open
Abstract
Obesity is considered a health hazard in part due to the associated multiple diseases. As rates of obesity continue to increase, a new strategy for its prevention and treatment is required. Compound-K, an active ingredient in ginseng, possesses antioxidant, anti-inflammatory, and anti-cancer properties. Although ginseng has used as various therapeutics, its potential ability to alleviate metabolic diseases by regulating adipocyte differentiation is still unknown. In this study, we found that CK treatment significantly inhibited lipid droplet and adipogenesis by downregulating the mRNA expression of C/ebpα, Ppar-γ, Fabp4, Srebp1, and adiponectin as well as protein levels of C/EBPα, PPAR-γ, and FABP4. CK also decreased the production of reactive oxygen species (ROS), while it increased endogeneous antioxidant enzymes such as catalase, glutathione peroxidase (GPx), glutathione reductase (GR), superoxide dismutase (SOD) 3 and SOD2. We observed that CK treatment suppressed the expression of cyclin-dependent kinase 1 (CDK1) and cyclin B1 during the mitotic clonal expansion (MCE) of adipocyte differentiation, and it arrested adipocytes at the G2/M stage due to the increased expression of p21 and p27. CK decreased the phosphorylation of extracellular signal-regulated kinase (ERK) and p38 and protein kinase B (AKT) in early-stage adipogenesis. In addition, the inhibition of adipogenesis by CK significantly increased the phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC). Interestingly, AMPK pharmacological inhibition with Dorsomorphin limited the effect of CK on suppressing PPAR-γ expression in differentiated 3T3-L1 cells. Our results suggest that CK exerts anti-adipogenic effects in 3T3-L1 cells through the activation of AMPK and inhibition of ERK/p38 and AKT signaling pathways.
Collapse
Affiliation(s)
- Jung-Mi Oh
- Department of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Korea
| | - Sungkun Chun
- Department of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Korea
- Institute of Medical Sciences, Jeonbuk National University Medical School, Jeonju 54907, Korea
- Research Institute for Endocrine Sciences, Jeonbuk National University Medical School, Jeonju 54907, Korea
| |
Collapse
|
20
|
Modulation of adipose inflammation by cellular retinoic acid-binding protein 1. Int J Obes (Lond) 2022; 46:1759-1769. [PMID: 35794192 PMCID: PMC9492549 DOI: 10.1038/s41366-022-01175-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 11/08/2022]
Abstract
Objectives Obesity, a metabolic syndrome, is known to be related to inflammation, especially adipose tissue inflammation. Cellular interactions within the expanded white adipose tissue (WAT) in obesity contribute to inflammation and studies have suggested that inflammation is triggered by inflamed adipocytes that recruit M1 macrophages into WAT. What causes accumulation of unhealthy adipocytes is an important topic of investigation. This study aims to understand the action of Cellular Retinoic Acid Binding Protein 1 (CRABP1) in WAT inflammation. Methods Eight weeks-old wild type (WT) and Crabp1 knockout (CKO) mice were fed with a normal diet (ND) or high-fat diet (HFD) for 8 weeks. Body weight and food intake were monitored. WATs and serum were collected for cellular and molecular analyses to determine affected signaling pathways. In cell culture studies, primary adipocyte differentiation and bone marrow-derived macrophages (BMDM) were used to examine adipocytes’ effects, mediated by CRABP1, in macrophage polarization. The 3T3L1-adipocyte was used to validate relevant signaling pathways. Results CKO mice developed an obese phenotype, more severely under high-fat diet (HFD) feeding. Further, CKO’s WAT exhibited a more severe inflammatory state as compared to wild type (WT) WAT, with a significantly expanded M1-like macrophage population. However, this was not caused by intrinsic defects of CKO macrophages. Rather, CKO adipocytes produced a significantly reduced level of adiponectin and had significantly lowered mitochondrial DNA content. CKO adipocyte-conditioned medium, compared to WT control, inhibited M2-like (CD206+) macrophage polarization. Mechanistically, defects in CKO adipocytes involved the ERK1/2 signaling pathway that could be modulated by CRABP1. Conclusions This study shows that CRABP1 plays a protective role against HFD-induced WAT inflammation through, in part, its regulation of adiponectin production and mitochondrial homeostasis in adipocytes, thereby modulating macrophage polarization in WAT to control its inflammatory potential.
Collapse
|
21
|
Meister BM, Hong SG, Shin J, Rath M, Sayoc J, Park JY. Healthy versus Unhealthy Adipose Tissue Expansion: the Role of Exercise. J Obes Metab Syndr 2022; 31:37-50. [PMID: 35283364 PMCID: PMC8987461 DOI: 10.7570/jomes21096] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 12/14/2022] Open
Abstract
Although the hallmark of obesity is the expansion of adipose tissue, not all adipose tissue expansion is the same. Expansion of healthy adipose tissue is accompanied by adequate capillary angiogenesis and mitochondria-centered metabolic integrity, whereas expansion of unhealthy adipose tissue is associated with capillary and mitochondrial derangement, resulting in deposition of immune cells (M1-stage macrophages) and excess production of pro-inflammatory cytokines. Accumulation of these dysfunctional adipose tissues has been linked to the development of obesity comorbidities, such as type 2 diabetes, hypertension, dyslipidemia, and cardiovascular disease, which are leading causes of human mortality and morbidity in modern society. Mechanistically, vascular rarefaction and mitochondrial incompetency (for example, low mitochondrial content, fragmented mitochondria, defective mitochondrial respiratory function, and excess production of mitochondrial reactive oxygen species) are frequently observed in adipose tissue of obese patients. Recent studies have demonstrated that exercise is a potent behavioral intervention for preventing and reducing obesity and other metabolic diseases. However, our understanding of potential cellular mechanisms of exercise, which promote healthy adipose tissue expansion, is at the beginning stage. In this review, we hypothesize that exercise can induce unique physiological stimuli that can alter angiogenesis and mitochondrial remodeling in adipose tissues and ultimately promote the development and progression of healthy adipogenesis. We summarize recent reports on how regular exercise can impose differential processes that lead to the formation of either healthy or unhealthy adipose tissue and discuss key knowledge gaps that warrant future research.
Collapse
Affiliation(s)
- Benjamin M Meister
- Department of Kinesiology, College of Public Health and Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Soon-Gook Hong
- Department of Kinesiology, College of Public Health and Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Junchul Shin
- Department of Kinesiology, College of Public Health and Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Meghan Rath
- Department of Kinesiology, College of Public Health and Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Jacqueline Sayoc
- Department of Kinesiology, College of Public Health and Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Joon-Young Park
- Department of Kinesiology, College of Public Health and Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
22
|
Van Meijel RLJ, Wang P, Bouwman F, Blaak EE, Mariman ECM, Goossens GH. The Effects of Mild Intermittent Hypoxia Exposure on the Abdominal Subcutaneous Adipose Tissue Proteome in Overweight and Obese Men: A First-in-Human Randomized, Single-Blind, and Cross-Over Study. Front Physiol 2022; 12:791588. [PMID: 35058800 PMCID: PMC8764283 DOI: 10.3389/fphys.2021.791588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Adipose tissue (AT) oxygen tension (pO2) has been implicated in AT dysfunction and metabolic perturbations in both rodents and humans. Compelling evidence suggests that hypoxia exposure alters metabolism, at least partly through effects on AT. However, it remains to be elucidated whether mild intermittent hypoxia (MIH) exposure impacts the AT proteome. We performed a randomized, single-blind, and cross-over study to investigate the effects of seven consecutive days of MIH (FiO2 15%, 3x2h/d) compared to normoxia (FiO2 21%) exposure on the AT proteome in overweight/obese men. In vivo AT insulin sensitivity was determined by the gold standard hyperinsulinemic-euglycemic clamp, and abdominal subcutaneous AT biopsies were collected under normoxic fasting conditions following both exposure regimens (day 8). AT proteins were isolated and quantified using liquid chromatography-mass spectrometry. After correction for blood contamination, 1,022 AT protein IDs were identified, of which 123 were differentially expressed following MIH (p < 0.05). We demonstrate for the first time that MIH exposure, which markedly reduces in vivo AT oxygen tension, impacts the human AT proteome. Although we cannot exclude that a single differentially expressed protein might be a false positive finding, several functional pathways were altered by MIH exposure, also after adjustment for multiple testing. Specifically, differentially expressed proteins were involved in redox systems, cell-adhesion, actin cytoskeleton organization, extracellular matrix composition, and energy metabolism. The MIH-induced change in AT TMOD3 expression was strongly related to altered in vivo AT insulin sensitivity, thus linking MIH-induced effects on the AT proteome to metabolic changes in overweight/obese humans.
Collapse
Affiliation(s)
- Rens L J Van Meijel
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Ping Wang
- Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Freek Bouwman
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Edwin C M Mariman
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
23
|
Gangwar A, Paul S, Arya A, Ahmad Y, Bhargava K. Altitude acclimatization via hypoxia-mediated oxidative eustress involves interplay of protein nitrosylation and carbonylation: A redoxomics perspective. Life Sci 2021; 296:120021. [PMID: 34626604 DOI: 10.1016/j.lfs.2021.120021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/22/2021] [Accepted: 09/30/2021] [Indexed: 12/17/2022]
Abstract
AIM Hypoxia is an important feature of multiple diseases like cancer and obesity and also an environmental stressor to high altitude travelers. Emerging research suggests the importance of redox signaling in physiological responses transforming the notion of oxidative stress into eustress and distress. However, the behavior of redox protein post-translational modifications (PTMs), and their correlation with stress acclimatization in humans remains sketchy. Scant information exists about modifications in redoxome during physiological exposure to environmental hypoxia. In this study, we investigated redox PTMs, nitrosylation and carbonylation, in context of extended environmental hypoxia exposure. METHODS The volunteers were confirmed to be free of any medical conditions and matched for age and weight. The human global redoxome and the affected networks were investigated using TMT-labeled quantitative proteo-bioinformatics and biochemical assays. The percolator PSM algorithm was used for peptide-spectrum match (PSM) validation in database searches. The FDR for peptide matches was set to 0.01. 1-way ANOVA and Tukey's Multiple Comparison test were used for biochemical assays. p-value<0.05 was considered statistically significant. Three independent experiments (biological replicates) were performed. Results were presented as Mean ± standard error of mean (SEM). KEY FINDINGS This investigation revealed direct and indirect interplay between nitrosylation and carbonylation especially within coagulation and inflammation networks; interlinked redox signaling (via nitrosylation‑carbonylation); and novel nitrosylation and carbonylation sites in individual proteins. SIGNIFICANCE This study elucidates the role of redox PTMs in hypoxia signaling favoring tolerance and survival. Also, we demonstrated direct and indirect interplay between nitrosylation and carbonylation is crucial to extended hypoxia tolerance.
Collapse
Affiliation(s)
- Anamika Gangwar
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India
| | - Subhojit Paul
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India
| | - Aditya Arya
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India
| | - Yasmin Ahmad
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India.
| | - Kalpana Bhargava
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India.
| |
Collapse
|
24
|
Jukić I, Kolobarić N, Stupin A, Matić A, Kozina N, Mihaljević Z, Mihalj M, Šušnjara P, Stupin M, Ćurić ŽB, Selthofer-Relatić K, Kibel A, Lukinac A, Kolar L, Kralik G, Kralik Z, Széchenyi A, Jozanović M, Galović O, Medvidović-Kosanović M, Drenjančević I. Carnosine, Small but Mighty-Prospect of Use as Functional Ingredient for Functional Food Formulation. Antioxidants (Basel) 2021; 10:1037. [PMID: 34203479 PMCID: PMC8300828 DOI: 10.3390/antiox10071037] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 11/17/2022] Open
Abstract
Carnosine is a dipeptide synthesized in the body from β-alanine and L-histidine. It is found in high concentrations in the brain, muscle, and gastrointestinal tissues of humans and is present in all vertebrates. Carnosine has a number of beneficial antioxidant properties. For example, carnosine scavenges reactive oxygen species (ROS) as well as alpha-beta unsaturated aldehydes created by peroxidation of fatty acid cell membranes during oxidative stress. Carnosine can oppose glycation, and it can chelate divalent metal ions. Carnosine alleviates diabetic nephropathy by protecting podocyte and mesangial cells, and can slow down aging. Its component, the amino acid beta-alanine, is particularly interesting as a dietary supplement for athletes because it increases muscle carnosine, and improves effectiveness of exercise and stimulation and contraction in muscles. Carnosine is widely used among athletes in the form of supplements, but rarely in the population of cardiovascular or diabetic patients. Much less is known, if any, about its potential use in enriched food. In the present review, we aimed to provide recent knowledge on carnosine properties and distribution, its metabolism (synthesis and degradation), and analytical methods for carnosine determination, since one of the difficulties is the measurement of carnosine concentration in human samples. Furthermore, the potential mechanisms of carnosine's biological effects in musculature, metabolism and on immunomodulation are discussed. Finally, this review provides a section on carnosine supplementation in the form of functional food and potential health benefits and up to the present, neglected clinical use of carnosine.
Collapse
Affiliation(s)
- Ivana Jukić
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia; (I.J.); (N.K.); (A.S.); (A.M.); (N.K.); (Z.M.); (M.M.); (P.Š.); (M.S.); (A.K.)
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
| | - Nikolina Kolobarić
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia; (I.J.); (N.K.); (A.S.); (A.M.); (N.K.); (Z.M.); (M.M.); (P.Š.); (M.S.); (A.K.)
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
| | - Ana Stupin
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia; (I.J.); (N.K.); (A.S.); (A.M.); (N.K.); (Z.M.); (M.M.); (P.Š.); (M.S.); (A.K.)
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Department of Pathophysiology, Physiology and Immunology, Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Cara Hadrijana 10E, HR-31000 Osijek, Croatia
| | - Anita Matić
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia; (I.J.); (N.K.); (A.S.); (A.M.); (N.K.); (Z.M.); (M.M.); (P.Š.); (M.S.); (A.K.)
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
| | - Nataša Kozina
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia; (I.J.); (N.K.); (A.S.); (A.M.); (N.K.); (Z.M.); (M.M.); (P.Š.); (M.S.); (A.K.)
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
| | - Zrinka Mihaljević
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia; (I.J.); (N.K.); (A.S.); (A.M.); (N.K.); (Z.M.); (M.M.); (P.Š.); (M.S.); (A.K.)
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
| | - Martina Mihalj
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia; (I.J.); (N.K.); (A.S.); (A.M.); (N.K.); (Z.M.); (M.M.); (P.Š.); (M.S.); (A.K.)
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Department of Dermatology and Venereology, University Hospital Osijek, HR-31000 Osijek, Croatia
| | - Petar Šušnjara
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia; (I.J.); (N.K.); (A.S.); (A.M.); (N.K.); (Z.M.); (M.M.); (P.Š.); (M.S.); (A.K.)
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
| | - Marko Stupin
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia; (I.J.); (N.K.); (A.S.); (A.M.); (N.K.); (Z.M.); (M.M.); (P.Š.); (M.S.); (A.K.)
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Department for Cardiovascular Disease, University Hospital Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
| | - Željka Breškić Ćurić
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Department of Internal Medicine, General Hospital Vinkovci, Zvonarska 57, HR-32100 Vinkovci, Croatia
| | - Kristina Selthofer-Relatić
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Department for Cardiovascular Disease, University Hospital Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
- Department for Internal Medicine, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
| | - Aleksandar Kibel
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia; (I.J.); (N.K.); (A.S.); (A.M.); (N.K.); (Z.M.); (M.M.); (P.Š.); (M.S.); (A.K.)
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Department for Cardiovascular Disease, University Hospital Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
| | - Anamarija Lukinac
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Department of Rheumatology, Clinical Immunology and Allergology, Clinical Hospital Center Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
| | - Luka Kolar
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Department of Internal Medicine, Vukovar General Hospital, HR-32000 Vukovar, Croatia
| | - Gordana Kralik
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Nutricin j.d.o.o. Darda, HR-31326 Darda, Croatia
| | - Zlata Kralik
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Department of Animal Production and Biotechnology, Faculty of Agrobiotechnical Sciences, Josip Juraj Strossmayer University of Osijek, Vladimira Preloga 1, HR-31000 Osijek, Croatia
| | - Aleksandar Széchenyi
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Department of Chemistry, Josip Juraj Strossmayer University of Osijek, Cara Hadrijana 8/A, HR-31000 Osijek, Croatia
| | - Marija Jozanović
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Department of Chemistry, Josip Juraj Strossmayer University of Osijek, Cara Hadrijana 8/A, HR-31000 Osijek, Croatia
| | - Olivera Galović
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Department of Chemistry, Josip Juraj Strossmayer University of Osijek, Cara Hadrijana 8/A, HR-31000 Osijek, Croatia
| | - Martina Medvidović-Kosanović
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
- Department of Chemistry, Josip Juraj Strossmayer University of Osijek, Cara Hadrijana 8/A, HR-31000 Osijek, Croatia
| | - Ines Drenjančević
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia; (I.J.); (N.K.); (A.S.); (A.M.); (N.K.); (Z.M.); (M.M.); (P.Š.); (M.S.); (A.K.)
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia; (Ž.B.Ć.); (K.S.-R.); (A.L.); (L.K.); (G.K.); (Z.K.); (A.S.); (M.J.); (O.G.); (M.M.-K.)
| |
Collapse
|
25
|
Kobayashi M, Deguchi Y, Nozaki Y, Higami Y. Contribution of PGC-1α to Obesity- and Caloric Restriction-Related Physiological Changes in White Adipose Tissue. Int J Mol Sci 2021; 22:ijms22116025. [PMID: 34199596 PMCID: PMC8199692 DOI: 10.3390/ijms22116025] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 12/16/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ coactivator-1 α (PGC-1α) regulates mitochondrial DNA replication and mitochondrial gene expression by interacting with several transcription factors. White adipose tissue (WAT) mainly comprises adipocytes that store triglycerides as an energy resource and secrete adipokines. The characteristics of WAT vary in response to systemic and chronic metabolic alterations, including obesity or caloric restriction. Despite a small amount of mitochondria in white adipocytes, accumulated evidence suggests that mitochondria are strongly related to adipocyte-specific functions, such as adipogenesis and lipogenesis, as well as oxidative metabolism for energy supply. Therefore, PGC-1α is expected to play an important role in WAT. In this review, we provide an overview of the involvement of mitochondria and PGC-1α with obesity- and caloric restriction-related physiological changes in adipocytes and WAT.
Collapse
Affiliation(s)
- Masaki Kobayashi
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Japan; (Y.D.); (Y.N.)
- Correspondence: (M.K.); (Y.H.); Tel.: +81-4-7121-3676 (M.K. & Y.H.)
| | - Yusuke Deguchi
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Japan; (Y.D.); (Y.N.)
| | - Yuka Nozaki
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Japan; (Y.D.); (Y.N.)
| | - Yoshikazu Higami
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda 278-8510, Japan; (Y.D.); (Y.N.)
- Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda 278-8510, Japan
- Correspondence: (M.K.); (Y.H.); Tel.: +81-4-7121-3676 (M.K. & Y.H.)
| |
Collapse
|
26
|
Zhou S, Zhang Q, Gao Y, Liu F, Cao W, Li Z, Xue C, Tang Q. Sargassum fusiforme together with turmeric extract and pomegranate peel extract alleviates obesity in high fat-fed C57BL/6J mice. Food Funct 2021; 12:4654-4669. [PMID: 33913445 DOI: 10.1039/d0fo03044a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Sargassum fusiforme, a nutritious edible brown alga, has been widely suggested to play an important role in the development of functional food because of its multiple biological activities. The aim of this study was to explore the anti-obesity effect of the combination of Sargassum fusiforme with extracts of fruit and vegetable by comparing the effects of Sargassum fusiforme (S), Sargassum fusiforme together with pomegranate peel extract (SP), Sargassum fusiforme together with turmeric extract (ST) and Sargassum fusiforme together with turmeric extract and pomegranate peel extract (C) on diet-induced obese C57BL/6J mice. Long-term consumption of a high-fat diet can lead to high levels of blood lipid, increase adipocyte size, and cause lipid metabolism dysfunction and gut microbiota dysbiosis. According to the results of the experiments, SP and ST were more effective in reducing lipid levels and fat accumulation than S; and, C exhibited the strongest efficacy compared with the other three supplements. ST and C also regulated adipocytokines and had significant effects on the gene expression of lipid metabolism. We also found that C alleviated the imbalance of intestinal flora caused by a high-fat diet to a certain extent. In conclusion, SP, ST and C have anti-obesity potentials, which can be used as alternative ingredients in the formula of functional food for obese people.
Collapse
Affiliation(s)
- Sainan Zhou
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266100, China.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Zhou Y, Li H, Xia N. The Interplay Between Adipose Tissue and Vasculature: Role of Oxidative Stress in Obesity. Front Cardiovasc Med 2021; 8:650214. [PMID: 33748199 PMCID: PMC7969519 DOI: 10.3389/fcvm.2021.650214] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/10/2021] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular diseases (CVDs) rank the leading cause of morbidity and mortality globally. Obesity and its related metabolic syndrome are well-established risk factors for CVDs. Therefore, understanding the pathophysiological role of adipose tissues is of great importance in maintaining cardiovascular health. Oxidative stress, characterized by excessive formation of reactive oxygen species, is a common cellular stress shared by obesity and CVDs. While plenty of literatures have illustrated the vascular oxidative stress, very few have discussed the impact of oxidative stress in adipose tissues. Adipose tissues can communicate with vascular systems, in an endocrine and paracrine manner, through secreting several adipocytokines, which is largely dysregulated in obesity. The aim of this review is to summarize current understanding of the relationship between oxidative stress in obesity and vascular endothelial dysfunction. In this review, we briefly describe the possible causes of oxidative stress in obesity, and the impact of obesity-induced oxidative stress on adipose tissue function. We also summarize the crosstalk between adipose tissue and vasculature mediated by adipocytokines in vascular oxidative stress. In addition, we highlight the potential target mediating adipose tissue oxidative stress.
Collapse
Affiliation(s)
- Yawen Zhou
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
28
|
The Effects of the Combination of Buckwheat D-Fagomine and Fish Omega-3 Fatty Acids on Oxidative Stress and Related Risk Factors in Pre-Obese Rats. Foods 2021; 10:foods10020332. [PMID: 33557198 PMCID: PMC7913974 DOI: 10.3390/foods10020332] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/18/2021] [Accepted: 02/01/2021] [Indexed: 11/16/2022] Open
Abstract
The combined supplementation of buckwheat D-fagomine (FG) and fish omega-3 polyunsaturated fatty acids (ω-3 PUFA) attenuates the development of insulin resistance in rats fed a high-fat (HF) diet. This study aimed to examine the effects of combined supplementation with FG and ω-3 PUFA on dyslipidemia, transaminases, interleukin-6, and oxidative stress. Forty-five male Sprague-Dawley rats were fed a standard diet, an HF diet, an HF diet supplemented with FG, an HF diet supplemented with ω-3 PUFA, or an HF diet supplemented with FG and ω-3 PUFA for 21 weeks. Triacylglycerol, cholesterol, aspartate aminotransferase, alanine aminotransferase, and interleukin-6 were measured. The assessment of oxidative stress included plasma antioxidant capacity, antioxidant enzyme activities, glutathione content, lipid peroxidation, and protein carbonylation. The combined supplementation with FG and ω-3 PUFA did not attenuate the slight accumulation of liver cholesterol induced by the HF diet but normalized the plasma alanine aminotransferase activity. Rats fed the HF diet supplemented with the combination showed a lower amount of plasma interleukin-6 than those fed a standard diet. The combination attenuated oxidative damage induced by the HF diet, decreased antioxidant enzyme activities, and enhanced glutathione status. The beneficial effects of the combination of FG and ω-3 PUFA on oxidative stress and related risk factors in pre-obese rats were mainly modulated by ω-3 PUFA.
Collapse
|
29
|
Tang Y, Zhao Y, Wang P, Sang S. Simultaneous Determination of Multiple Reactive Carbonyl Species in High Fat Diet-Induced Metabolic Disordered Mice and the Inhibitory Effects of Rosemary on Carbonyl Stress. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:1123-1131. [PMID: 33464893 DOI: 10.1021/acs.jafc.0c07748] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
As potential endogenous biomarkers, reactive carbonyl species (RCS) have gained abundant attention for monitoring oxidative and carbonyl stress. However, there is no accurate method to evaluate multiple RCS in biological samples. In this study, a 2,4-dinitrophenylhydrazine (DNPH) derivatization-based LC-MS method was developed and validated to quantitate eight RCS: malondialdehyde (MDA), acrolein (ACR), 4-hydroxy-2-nonenal (4-HNE), 4-oxo-2-nonenal (4-ONE), methylglyoxal (MGO), glyoxal (GO), 3-deoxyglucosone (3-DG), and 2-keto-d-glucose (2-Keto). Subsequently, the method was applied to assess the RCS in low fat (LF), high fat (HF), and HF plus rosemary extract (RE) diet-fed mouse samples. The quantitative results on RCS levels indicated that the HF diet significantly increased the total RCS levels in mouse urine, plasma, and kidney with an average rate of 280.69%, 153.87%, and 61.30%, respectively. The RE administration significantly inhibited the elevated RCS levels induced by the HF diet, especially for MDA, 4-ONE, 4-HNE, and 2-Keto in mouse plasma, and ACR and 2-Keto in mouse kidney. This is the first study to simultaneously measure eight RCS in biological samples and demonstrate that RE was able to eliminate the accumulation of the HF diet-induced RCS.
Collapse
Affiliation(s)
- Yao Tang
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, 500 Laureate Way, Kannapolis, North Carolina 28081, United States
| | - Yantao Zhao
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, 500 Laureate Way, Kannapolis, North Carolina 28081, United States
| | - Pei Wang
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, 500 Laureate Way, Kannapolis, North Carolina 28081, United States
| | - Shengmin Sang
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, 500 Laureate Way, Kannapolis, North Carolina 28081, United States
| |
Collapse
|
30
|
Hauck AK, Zhou T, Upadhyay A, Sun Y, O’Connor MB, Chen Y, Bernlohr DA. Histone Carbonylation Is a Redox-Regulated Epigenomic Mark That Accumulates with Obesity and Aging. Antioxidants (Basel) 2020; 9:antiox9121210. [PMID: 33271806 PMCID: PMC7761391 DOI: 10.3390/antiox9121210] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress is a hallmark of metabolic disease, though the mechanisms that define this link are not fully understood. Irreversible modification of proteins by reactive lipid aldehydes (protein carbonylation) is a major consequence of oxidative stress in adipose tissue and the substrates and specificity of this modification are largely unexplored. Here we show that histones are avidly modified by 4-hydroxynonenal (4-HNE) in vitro and in vivo. Carbonylation of histones by 4-HNE increased with age in male flies and visceral fat depots of mice and was potentiated in genetic (ob/ob) and high-fat feeding models of obesity. Proteomic evaluation of in vitro 4-HNE- modified histones led to the identification of both Michael and Schiff base adducts. In contrast, mapping of sites in vivo from obese mice exclusively revealed Michael adducts. In total, we identified 11 sites of 4-hydroxy hexenal (4-HHE) and 10 sites of 4-HNE histone modification in visceral adipose tissue. In summary, these results characterize adipose histone carbonylation as a redox-linked epigenomic mark associated with metabolic disease and aging.
Collapse
Affiliation(s)
- Amy K. Hauck
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; (A.K.H.); (T.Z.); (Y.C.)
| | - Tong Zhou
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; (A.K.H.); (T.Z.); (Y.C.)
| | - Ambuj Upadhyay
- Department of Molecular Biology, Cell Biology, Developmental Biology and Genetics, University of Minnesota, Minneapolis, MN 55455, USA; (A.U.); (M.B.O.)
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA;
| | - Michael B. O’Connor
- Department of Molecular Biology, Cell Biology, Developmental Biology and Genetics, University of Minnesota, Minneapolis, MN 55455, USA; (A.U.); (M.B.O.)
| | - Yue Chen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; (A.K.H.); (T.Z.); (Y.C.)
| | - David A. Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; (A.K.H.); (T.Z.); (Y.C.)
- Correspondence:
| |
Collapse
|
31
|
Man AWC, Xia N, Li H. Circadian Rhythm in Adipose Tissue: Novel Antioxidant Target for Metabolic and Cardiovascular Diseases. Antioxidants (Basel) 2020; 9:E968. [PMID: 33050331 PMCID: PMC7601443 DOI: 10.3390/antiox9100968] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/29/2020] [Accepted: 10/03/2020] [Indexed: 12/11/2022] Open
Abstract
Obesity is a major risk factor for most metabolic and cardiovascular disorders. Adipose tissue is an important endocrine organ that modulates metabolic and cardiovascular health by secreting signaling molecules. Oxidative stress is a common mechanism associated with metabolic and cardiovascular complications including obesity, type 2 diabetes, and hypertension. Oxidative stress can cause adipose tissue dysfunction. Accumulating data from both humans and experimental animal models suggest that adipose tissue function and oxidative stress have an innate connection with the intrinsic biological clock. Circadian clock orchestrates biological processes in adjusting to daily environmental changes according to internal or external cues. Recent studies have identified the genes and molecular pathways exhibiting circadian expression patterns in adipose tissue. Disruption of the circadian rhythmicity has been suggested to augment oxidative stress and aberrate adipose tissue function and metabolism. Therefore, circadian machinery in the adipose tissue may be a novel therapeutic target for the prevention and treatment of metabolic and cardiovascular diseases. In this review, we summarize recent findings on circadian rhythm and oxidative stress in adipose tissue, dissect the key components that play a role in regulating the clock rhythm, oxidative stress and adipose tissue function, and discuss the potential use of antioxidant treatment on metabolic and cardiovascular diseases by targeting the adipose clock.
Collapse
Affiliation(s)
| | | | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr, 1, 55131 Mainz, Germany; (A.W.C.M.); (N.X.)
| |
Collapse
|
32
|
Magalhães SC, de Oliveira KA, Freiras PA, Moreira Gomes MD, Pereira LM, Boa LF, de Carvalho DP, Fortunato RS, Carneiro Loureiro AC, Brito LC, de Oliveira AC. High-dose Nandrolone Decanoate induces oxidative stress and inflammation in retroperitoneal adipose tissue of male rats. J Steroid Biochem Mol Biol 2020; 203:105728. [PMID: 32712213 DOI: 10.1016/j.jsbmb.2020.105728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/06/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023]
Abstract
The non-therapeutic use of the androgenic anabolic steroid Nandrolone Decanoate is popular due to its effects on physical performance and body composition, especially for its lipolytic and anabolic effects associated. However, high doses of such drugs are often associated with a series of pathologies related to unbalanced redox homeostasis, which, in turn, can be linked to inflammation. The oxidative stress onset could deregulate the secretion of cytokines, evidencing a dysfunctional adipocyte. Thus, the aim of this study was to investigate the effect of supraphysiological doses of Nandrolone Decanoate on redox homeostasis of retroperitoneal fatpad of male rats and its relationship with cytokines-based inflammatory signaling. Hydrogen peroxide production was assessed in the retroperitoneal fat pad of adult male rats which received either 10 mg kg of Nandrolone Decanoate or only a vehicle. Also, catalase, superoxide dismutase and glutathione peroxidase activities were measured, together with total reduced thiols and protein carbonylation, as well as IL-1β, TNF-α, and IL-6 local levels. High doses of Nandrolone Decanoate caused an increase in the hydrogen peroxide production, together with lower activities of the antioxidant enzymes and lower levels of total reduced thiol. There were also higher protein carbonylation and greater levels of IL-1β, TNF-α, and IL-6 in the treated group compared to control group. Therefore, it was possible to verify that high doses of Nandrolone Decanoate cause oxidative stress and induce higher inflammatory signaling in retroperitoneal fat pad of male rats.
Collapse
Affiliation(s)
- Saulo Chaves Magalhães
- Laboratório de Fisiologia Endócrina e Metabolismo, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Ceará, Brazil
| | - Keciany Alves de Oliveira
- Laboratório de Fisiologia Endócrina e Metabolismo, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Ceará, Brazil
| | - Paula Alexandre Freiras
- Laboratório de Fisiologia Endócrina e Metabolismo, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Ceará, Brazil
| | - Maria Diana Moreira Gomes
- Laboratório de Eletrofisiologia, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Ceará, Brazil
| | - Leonardo Matta Pereira
- Laboratório de Eletrofisiologia, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Ceará, Brazil
| | - Luiz Fonte Boa
- Laboratório de Eletrofisiologia, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Ceará, Brazil
| | - Denise Pires de Carvalho
- Laboratório de Fisiologia Endócrina Dóris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo Soares Fortunato
- Laboratório de Fisiologia e Sinalização Redox, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adriano Cesar Carneiro Loureiro
- Laboratório de Bioquímica e Expressão Gênica, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Ceará, Brazil
| | - Luciana Catunda Brito
- Instututo de Educação Física e Esportes, Universidade Federal do Ceará, Ceará, Brazil
| | - Ariclécio Cunha de Oliveira
- Laboratório de Fisiologia Endócrina e Metabolismo, Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Ceará, Brazil.
| |
Collapse
|
33
|
Caimi G, Hopps E, Montana M, Carollo C, Calandrino V, Gallà E, Canino B, Lo Presti R. Behaviour of carbonyl groups in several clinical conditions: Analysis of our survey. Clin Hemorheol Microcirc 2020; 74:299-313. [PMID: 31683469 DOI: 10.3233/ch-190689] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Protein carbonylation is a marker of oxidative protein damage, that is likely involved in the pathogenesis of several diseases. The aim of this study was to evaluate the protein carbonyl (PC) groups in different clinical conditions. It included different groups of subjects: 81 trained subjects; 23 subjects with mild essential hypertension; 31 middle-aged subjects with metabolic syndrome (MS); 106 subjects with MS not selected for age (subdivided into two subgroups, with and without diabetes mellitus); 91 obese adults subdivided in two subgroups (BMI 30-35 Kg/m2 and BMI > 35 kg/m2); 48 subjects with obstructive sleep apnea syndrome (OSAS) subdivided in accordance with the apnea/hypopnea index (AHI); 27 subjects with chronic kidney disease (CKD) on conservative therapy; 31 subjects with CKD on haemodialysis treatment; and 50 subjects with juvenile myocardial infarction. PC groups were reduced in trained subjects in comparison with sedentary controls, while no variation was observed in mild essential hypertension. PC groups were increased in MS subjects and in adult obese subjects. In MS subjects the PC groups were not influenced by the presence of diabetes mellitus and in adult obese subjects were not influenced by the obesity degree. In OSAS subjects only those with AHI > 30 showed an increase of PC groups. PC groups increased in CKD subjects undergoing conservative treatment and haemodialysis therapy. In dialyzed subjects, after a standard dialysis session, there was a marked increase in PC groups. In juvenile myocardial infarction PC groups were higher than in controls; there was no difference between STEMI and NSTEMI and their concentration was unaffected by the number of cardiovascular risk factors or stenosed coronary vessels.
Collapse
Affiliation(s)
- Gregorio Caimi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Università degli Studi di Palermo, Palermo, Italy
| | - Eugenia Hopps
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Università degli Studi di Palermo, Palermo, Italy
| | - Maria Montana
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Università degli Studi di Palermo, Palermo, Italy
| | - Caterina Carollo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Università degli Studi di Palermo, Palermo, Italy
| | - Vincenzo Calandrino
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Università degli Studi di Palermo, Palermo, Italy
| | - Eleonora Gallà
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Università degli Studi di Palermo, Palermo, Italy
| | - Baldassare Canino
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Università degli Studi di Palermo, Palermo, Italy
| | - Rosalia Lo Presti
- Department of Psychology, Educational Science and Human Movement, Università degli Studi di Palermo, Palermo, Italy
| |
Collapse
|
34
|
Kim J, Kim JS, Kim SS, Jung JG, Yoon SJ, Seo YR, Lee S, Bae YK, Lee WJ. Influence of Facial Flushing on Pre- or Type 2 Diabetes Risk according to Alcohol Consumption in Korean Male. Korean J Fam Med 2020; 41:153-160. [PMID: 32456383 PMCID: PMC7272365 DOI: 10.4082/kjfm.18.0120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/14/2018] [Indexed: 01/29/2023] Open
Abstract
Background This study aims to examine the association between alcohol consumption and the risk of pre- or type 2 diabetes mellitus (T2DM) by alcohol-induced flushing response in Korean male adults, particularly based on their body mass index (BMI). Methods This study selected 1,030 (158 non-drinkers, 364 flushers, and 508 non-flushers) male adults who had medical checkups. A logistic regression analysis was used to compare the association between alcohol consumption and the risk of pre- or T2DM. Results In both the normal-weight group (BMI <23 kg/m2) and the overweight group (BMI ≥23 kg/m2 and <25 kg/ m2), the flushers had a higher risk of pre- or T2DM (odds ratio, 95% confidence interval) when consuming more than 8 drinks of alcohol per week than the non-drinkers (normal-weight group: 3.43, 1.06–11.07; overweight group: 4.94, 1.56–15.67). But in the non-flushers among the normal-weight group and the overweight group, there was no significant difference compared to non-drinkers regarding the risk of pre- or T2DM. Obese flushers had a significantly higher risk of pre- or T2DM when consuming more than 4 drinks of alcohol per week than the non-drinkers (>4 and ≤8 drinks: 2.64, 1.10–6.36; >8 drinks: 2.42, 1.11–5.27). However, obese non-flushers had only a significant higher risk of pre- or T2DM when consuming more than 8 drinks of alcohol per week than the non-drinkers (2.72, 1.39–5.30) Conclusion These results suggest that obese flushers have an increased risk of developing pre- or T2DM even with less alcohol consumption.
Collapse
Affiliation(s)
- Jihan Kim
- Department of Family Medicine, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jong Sung Kim
- Department of Family Medicine, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Sung-Soo Kim
- Department of Family Medicine, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jin-Gyu Jung
- Department of Family Medicine, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Seok-Jun Yoon
- Department of Family Medicine, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Yu-Ri Seo
- Department of Family Medicine, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Sami Lee
- Department of Family Medicine, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Yoon-Kyung Bae
- Department of Family Medicine, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Won-Jin Lee
- Department of Family Medicine, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| |
Collapse
|
35
|
Manuel AM, Walla MD, Dorn MT, Tanis RM, Piroli GG, Frizzell N. Fumarate and oxidative stress synergize to promote stability of C/EBP homologous protein in the adipocyte. Free Radic Biol Med 2020; 148:70-82. [PMID: 31883977 PMCID: PMC6961135 DOI: 10.1016/j.freeradbiomed.2019.12.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/22/2019] [Accepted: 12/24/2019] [Indexed: 12/13/2022]
Abstract
C/EBP homologous protein (CHOP) is a transcription factor that is elevated in adipose tissue across many models of diabetes and metabolic stress. Although increased CHOP levels are associated with the terminal response to endoplasmic reticulum stress and apoptosis, there is no evidence for CHOP mediated apoptosis in the adipose tissue during diabetes. CHOP protein levels increase in parallel with protein succination, a fumarate derived cysteine modification, in the adipocyte during metabolic stress. We investigated the factors contributing to sustained CHOP proteins levels in the adipocyte, with an emphasis on the regulation of CHOP protein turnover by metabolite-driven modification of Keap1 cysteines. CHOP protein stability was investigated in conditions of nutrient stress due to high glucose or elevated fumarate (fumarase knockdown model); where cysteine succination is specifically elevated. CHOP protein turnover is significantly reduced in models of elevated glucose and fumarate with a ~30% increase in CHOP stability (p > 0.01), in part due to decreased CHOP phosphorylation. Sustained CHOP levels occur in parallel with elevated heme-oxygenase-1, a production of increased Nrf2 transcriptional activity and Keap1 modification. While Keap1 is directly succinated in the presence of excess fumarate derived from genetic knockdown of fumarase (fumarate levels are elevated >20-fold), it is the oxidative modification of Keap1 that predominates in adipocytes matured in high glucose (fumarate increases 4-5 fold). Elevated fumarate indirectly regulates CHOP stability through the induction of oxidative stress. The antioxidant N-acetylcysteine (NAC) reduces fumarate levels, protein succination and CHOP levels in adipocytes matured in high glucose. Elevated CHOP does not contribute elevated apoptosis in adipocytes, but plays a redox-dependent role in decreasing the adipocyte secretion of interleukin-13, an anti-inflammatory chemokine. NAC treatment restores adipocyte IL-13 secretion, confirming the redox-dependent regulation of a potent anti-inflammatory eotaxin. This study demonstrates that physiological increases in the metabolite fumarate during high glucose exposure contributes to the presence of oxidative stress and sustained CHOP levels in the adipocyte during diabetes. The results reveal a novel metabolic link between mitochondrial metabolic stress and reduced anti-inflammatory adipocyte signaling as a consequence of reduced CHOP protein turnover.
Collapse
Affiliation(s)
- Allison M Manuel
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, 29209, USA
| | - Michael D Walla
- Mass Spectrometry Center, Department of Chemistry & Biochemistry, University of South Carolina, Columbia, SC, 29205, USA
| | - Margaret T Dorn
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, 29209, USA
| | - Ross M Tanis
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, 29209, USA
| | - Gerardo G Piroli
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, 29209, USA
| | - Norma Frizzell
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC, 29209, USA.
| |
Collapse
|
36
|
Masschelin PM, Cox AR, Chernis N, Hartig SM. The Impact of Oxidative Stress on Adipose Tissue Energy Balance. Front Physiol 2020; 10:1638. [PMID: 32038305 PMCID: PMC6987041 DOI: 10.3389/fphys.2019.01638] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 12/30/2019] [Indexed: 12/21/2022] Open
Abstract
Overnutrition and sedentary activity reinforce the growing trend of worldwide obesity, insulin resistance, and type 2 diabetes. However, we have limited insight into how food intake generates sophisticated metabolic perturbations associated with obesity. Accumulation of mitochondrial oxidative stress contributes to the metabolic changes in obesity, but the mechanisms and significance are unclear. In white adipose tissue (WAT), mitochondrial oxidative stress, and the generation of reactive oxygen species (ROS) impact the endocrine and metabolic function of fat cells. The central role of mitochondria in nutrient handling suggests pharmacological targeting of pathological oxidative stress likely improves the metabolic profile of obesity. This review will summarize the critical pathogenic mechanisms of obesity-driven oxidative stress in WAT.
Collapse
Affiliation(s)
- Peter M Masschelin
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, United States.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Aaron R Cox
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Natasha Chernis
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Sean M Hartig
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, United States.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
37
|
Monroe TB, Anderson EJ. A Highly Sensitive, Reproducible Assay for Determining 4-hydroxynonenal Protein Adducts in Biological Material. Bio Protoc 2019; 9:e3383. [PMID: 33654879 PMCID: PMC7853934 DOI: 10.21769/bioprotoc.3383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/04/2019] [Indexed: 11/02/2022] Open
Abstract
Oxidative stress is associated with numerous diseases, and markers of oxidative stress in biological material are becoming a mainstay of both experimental and clinical/epidemiological research. Lipid peroxidation is a major form of oxidative stress, but due to their rapid degradation and instability, lipid peroxides are notoriously difficult to measure, particularly in biological specimens where their production and removal are continuously occuring. Thus, a commonly used surrogate marker of lipid peroxidation is protein adducts of 4-Hydroxynonenal (HNE), an α, β-unsaturated hydroxyalkenal (i.e., a reactive aldehyde) formed via degradation of oxidized polyunsaturated fatty acids (PUFAs). HNE adducts can be measured via commercially-available immunosorbent assays, but these have their limitations due to excessive costs, and reproducibility among laboratories is challenging due to variability in assay sensitivity, procedure, and reagents. Here we present a reproducible, facile, and economically conservative protocol for quantifying HNE protein adducts. The key to this protocol is to generate HNE-adduct standards by incubating bovine serum albumin (BSA) with HNE. These standards are then adsorbed to immunsorbent plastic in a multi-well plate format alongside biological samples. An enzyme-linked immunosorbent assay (ELISA) is then performed on the multi-well plate using commercially-available primary and secondary antibodies, and a peroxide-based fluorescent developing reagent. This protocol is highly sensitive and offers advantages to commercial sources in that it allows for reproducible, high-throughput quantitation of HNE adducts in a large number of samples. As such, it may be useful as a biomarker of chronic oxidative stress for experimental and clinical studies.
Collapse
Affiliation(s)
- T. Blake Monroe
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy; University of Iowa, Iowa City, IA, USA
| | - Ethan J. Anderson
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy; University of Iowa, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
38
|
Serrano JCE, Baena‐Fustegueras JA, Martin‐Gari M, Rassendren H, Cassanye A, Naudí A, López‐Cano C, Sánchez E, de la Fuente‐Juárez MC, Herrerías González F, Olsina Kissler JJ, Lecube A, Portero‐Otín M. Adipose Tissue Protein Glycoxidation is Associated with Weight-Loss Potential. Obesity (Silver Spring) 2019; 27:1133-1140. [PMID: 31112015 PMCID: PMC6618070 DOI: 10.1002/oby.22501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/19/2019] [Indexed: 11/11/2022]
Abstract
OBJECTIVE This study aimed to characterize the differences in protein oxidation biomarkers in adipose tissue (AT) as an indicator of AT metabolism and bariatric surgery weight-loss success. METHODS A human model, in which sixty-five individuals with obesity underwent bariatric surgery, and a diet-induced obesity animal model, in which animals were treated for 2 months with normocaloric diets, were analyzed to determine the associations between AT protein oxidation and body weight loss. Protein oxidative biomarkers were determined by gas chromatography/mass spectrometry in AT from human volunteers before the surgery, as well as 2 months after a diet treatment in the animal model. RESULTS The levels of carboxyethyl-lysine (CEL) and 2-succinocystein (2SC) in both visceral and subcutaneous AT before the surgery directly correlated with greater weight loss in both human and animal models. 2SC levels in subcutaneous AT greater than 4.7 × 106 μmol/mol lysine (95% CI: 3.4 × 106 to 6.0 × 106 ) may predict greater weight loss after bariatric surgery (receiver operating characteristic curve area = 0.8222; P = 0.0047). Additionally, it was observed that individuals with diabetes presented lower levels of CEL and 2SC in subcutaneous AT (P = 0.0266 and P = 0.0316, respectively) compared with individuals without diabetes. CONCLUSIONS CEL and 2SC in AT are useful biomarkers of AT metabolism and predict the individual's ability to reduce body weight after bariatric surgery.
Collapse
Affiliation(s)
| | - Juan Antonio Baena‐Fustegueras
- General and Digestive Surgery Service, Endocrine, Bariatric and Metabolic Surgery UnitArnau de Vilanova University HospitalLleidaSpain
| | | | | | - Anna Cassanye
- Department of Experimental MedicineUniversity of LleidaLleidaSpain
| | - Alba Naudí
- Department of Experimental MedicineUniversity of LleidaLleidaSpain
| | - Carolina López‐Cano
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital; and Obesity, Diabetes and Metabolism Research Group (ODIM), Institut de Recerca Biomèdica de Lleida (IRBLleida)University of LleidaLleidaSpain
| | - Enric Sánchez
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital; and Obesity, Diabetes and Metabolism Research Group (ODIM), Institut de Recerca Biomèdica de Lleida (IRBLleida)University of LleidaLleidaSpain
| | - María Cruz de la Fuente‐Juárez
- General and Digestive Surgery Service, Endocrine, Bariatric and Metabolic Surgery UnitArnau de Vilanova University HospitalLleidaSpain
| | - Fernando Herrerías González
- General and Digestive Surgery Service, Endocrine, Bariatric and Metabolic Surgery UnitArnau de Vilanova University HospitalLleidaSpain
| | - Jorge J. Olsina Kissler
- General and Digestive Surgery Service, Endocrine, Bariatric and Metabolic Surgery UnitArnau de Vilanova University HospitalLleidaSpain
| | - Albert Lecube
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital; and Obesity, Diabetes and Metabolism Research Group (ODIM), Institut de Recerca Biomèdica de Lleida (IRBLleida)University of LleidaLleidaSpain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)Instituto de Salud Carlos III (ISCIII)MadridSpain
| | | |
Collapse
|
39
|
Abstract
Background Thermogenic adipocytes reorganize their metabolism during cold exposure. Metabolic reprogramming requires readily available bioenergetics substrates, such as glucose and fatty acids, to increase mitochondrial respiration and produce heat via the uncoupling protein 1 (UCP1). This condition generates a finely-tuned production of mitochondrial reactive oxygen species (ROS) that support non-shivering thermogenesis. Scope of review Herein, the findings underlining the mechanisms that regulate ROS production and control of the adaptive responses tuning thermogenesis in adipocytes are described. Furthermore, this review describes the metabolic responses to substrate availability and the consequence of mitochondrial failure to switch fuel oxidation in response to changes in nutrient availability. A framework to control mitochondrial ROS threshold to maximize non-shivering thermogenesis in adipocytes is provided. Major conclusions Thermogenesis synchronizes fuel oxidation with an acute and transient increase of mitochondrial ROS that promotes the activation of redox-sensitive thermogenic signaling cascade and UCP1. However, an overload of substrate flux to mitochondria causes a massive and damaging mitochondrial ROS production that affects mitochondrial flexibility. Finding novel thermogenic redox targets and manipulating ROS concentration in adipocytes appears to be a promising avenue of research for improving thermogenesis and counteracting metabolic diseases. Mitochondrial ROS support non-shivering thermogenesis. Thermogenic ROS are tightly related to mitochondrial metabolic reorganization. Uncontrolled mitochondrial ROS production is causative of metabolic inflexibility.
Collapse
|
40
|
Ben Lamine J, Boujbiha MA, Dahane S, Cherifa AB, Khlifi A, Chahdoura H, Yakoubi MT, Ferchichi S, El Ayeb N, Achour L. α-Amylase and α-glucosidase inhibitor effects and pancreatic response to diabetes mellitus on Wistar rats of Ephedra alata areal part decoction with immunohistochemical analyses. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:9739-9754. [PMID: 30729433 DOI: 10.1007/s11356-019-04339-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 01/22/2019] [Indexed: 06/09/2023]
Abstract
Ephedra alata, known as a medicinal plant in China, was used in this study as aqueous extract from aerial parts, for diabetes mellitus treatment. This study was carried out on two parts, in vitro, we tested the effect of the studied extract on the inhibition of α-glucosidase and α-amylase activities, and in vivo on Wistar male rats receiving alloxan intraperitoneally at a rate of 125 mg/kg. Extract (100, 200, and 300 mg/kg of body weight) was administrated for 28 days by oral gavage. Blood glucose, amylase, lipase, and lipid profile level were determined. Oxidative stress was evaluated by enzymatic activities of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx), and by estimation of lipid peroxidation and protein carbonyl (PC) level. Histopathological changes in pancreas were investigated under photonic microscopy using immunohistochemical procedure. Our findings showed that aqueous extract inhibited in vitro both α-glucosidase and α-amylase activities and its use in vivo at 300 mg/kg of body weight restored pancreas weight and weight gain, ameliorated significantly (p ˂ 0.05) biochemical parameters; it prevented the increase in lipid and protein oxidation and the decrease in enzymatic and non-enzymatic defense system. Histological study of treated animals showed a comparable healed regeneration of beta cells.
Collapse
Affiliation(s)
- Jihene Ben Lamine
- Institut Supérieur de Biotechnologie de Monastir, Laboratoire de Recherche : Bioressources, Biologie Intégrative & Valorisation, Université de Monastir, LR14ES06, BP 74, 5000, Monastir, Tunisia.
- Faculté des Sciences de Tunis, Université de Tunis El Manar, 2092, Tunis, Tunisia.
| | - Mouhamed Ali Boujbiha
- Institut Supérieur de Biotechnologie de Monastir, Laboratoire de Recherche : Bioressources, Biologie Intégrative & Valorisation, Université de Monastir, LR14ES06, BP 74, 5000, Monastir, Tunisia
| | - Sabra Dahane
- Institut Supérieur de Biotechnologie de Monastir, Laboratoire de Recherche : Bioressources, Biologie Intégrative & Valorisation, Université de Monastir, LR14ES06, BP 74, 5000, Monastir, Tunisia
| | - Amal Ben Cherifa
- Institut Supérieur de Biotechnologie de Monastir, Laboratoire de Recherche : Bioressources, Biologie Intégrative & Valorisation, Université de Monastir, LR14ES06, BP 74, 5000, Monastir, Tunisia
- Faculté des Sciences de Gabes, Université de Gabes, Gabes, Tunisia
| | - Aida Khlifi
- Institut Supérieur de Biotechnologie de Monastir, Laboratoire de Recherche : Bioressources, Biologie Intégrative & Valorisation, Université de Monastir, LR14ES06, BP 74, 5000, Monastir, Tunisia
| | - Hassiba Chahdoura
- Institut Supérieur de Biotechnologie de Monastir, Laboratoire de Recherche : Bioressources, Biologie Intégrative & Valorisation, Université de Monastir, LR14ES06, BP 74, 5000, Monastir, Tunisia
| | - Mouhamed Taher Yakoubi
- Laboratoire d'anatomie et pathologie, Centre Hôpital Universitaire Farhat Hached, Sousse, Tunisia
| | - Salima Ferchichi
- Laboratoire de biochimie, Centre Hôpital Universitaire Farhat Hached, Sousse, Tunisia
| | - Nacer El Ayeb
- Institut Supérieur de Biotechnologie de Monastir, Laboratoire de Recherche : Bioressources, Biologie Intégrative & Valorisation, Université de Monastir, LR14ES06, BP 74, 5000, Monastir, Tunisia
| | - Lotfi Achour
- Institut Supérieur de Biotechnologie de Monastir, Laboratoire de Recherche : Bioressources, Biologie Intégrative & Valorisation, Université de Monastir, LR14ES06, BP 74, 5000, Monastir, Tunisia
| |
Collapse
|
41
|
Hauck AK, Huang Y, Hertzel AV, Bernlohr DA. Adipose oxidative stress and protein carbonylation. J Biol Chem 2018; 294:1083-1088. [PMID: 30563836 DOI: 10.1074/jbc.r118.003214] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Increased oxidative stress and abundance of reactive oxygen species (ROS) are positively correlated with a variety of pathophysiologies, including cardiovascular disease, type 2 diabetes, Alzheimer's disease, and neuroinflammation. In adipose biology, diabetic obesity is correlated with increased ROS in an age- and depot-specific manner and is mechanistically linked to mitochondrial dysfunction, endoplasmic reticulum (ER) stress, potentiated lipolysis, and insulin resistance. The cellular quality control systems that homeostatically regulate oxidative stress in the lean state are down-regulated in obesity as a consequence of inflammatory cytokine pressure leading to the accumulation of oxidized biomolecules. New findings have linked protein, DNA, and lipid oxidation at the biochemical level, and the structures and potential functions of protein adducts such as carbonylation that accumulate in stressed cells have been characterized. The sum total of such regulation and biochemical changes results in alteration of cellular metabolism and function in the obese state relative to the lean state and underlies metabolic disease progression. In this review, we discuss the molecular mechanisms and events underlying these processes and their implications for human health and disease.
Collapse
Affiliation(s)
- Amy K Hauck
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Yimao Huang
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455
| | - Ann V Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455.
| |
Collapse
|
42
|
Grun LK, Teixeira NDR, Mengden LV, de Bastiani MA, Parisi MM, Bortolin R, Lavandoski P, Pierdoná V, Alves LB, Moreira JCF, Mottin CC, Jones MH, Klamt F, Padoin AV, Guma FCR, Barbé-Tuana FM. TRF1 as a major contributor for telomeres' shortening in the context of obesity. Free Radic Biol Med 2018; 129:286-295. [PMID: 30268887 DOI: 10.1016/j.freeradbiomed.2018.09.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 09/24/2018] [Indexed: 01/02/2023]
Abstract
Obesity is a prevalent multifactorial chronic disorder characterized by metabolic dysregulation. Sustained pro-oxidative mediators trigger harmful consequences that reflect at systemic level and contribute for the establishment of a premature senescent phenotype associated with macromolecular damage (DNA, protein, and lipids). Telomeres are structures that protect chromosome ends and are associated with a six-protein complex called the shelterin complex and subject to regulation. Under pro-oxidant conditions, telomere attrition and the altered expression of the shelterin proteins are central for the establishment of many pathophysiological conditions such as obesity. Thus, considering that individuals with obesity display a systemic oxidative stress profile that may compromise the telomeres length or its regulation, the aim of this study was to investigate telomere homeostasis in patients with obesity and explore broad/systemic associations with the expression of shelterin genes and the plasma redox state. We performed a cross-sectional study in 39 patients with obesity and 27 eutrophic subjects. Telomere length (T/S ratio) and gene expression of shelterin components were performed in peripheral blood mononuclear cells by qPCR. The oxidative damage (lipid peroxidation and protein carbonylation) and non-enzymatic antioxidant system (total radical-trapping antioxidant potential/reactivity, sulfhydryl and GSH content) were evaluated in plasma. Our results demonstrate that independently of comorbidities, individuals with obesity had significantly shorter telomeres, augmented expression of negative regulators of the shelterin complex, increased lipid peroxidation and higher oxidized protein levels associated with increased non-enzymatic antioxidant defenses. Principal component analysis revealed TRF1 as a major contributor for firstly telomeres shortening. In conclusion, our study is first showing a comprehensive analysis of telomeres in the context of obesity, associated with dysregulation of the shelterin components that was partially explained by TRF1 upregulation that could not be reversed by the observed adaptive non-enzymatic antioxidant response.
Collapse
Affiliation(s)
- Lucas Kich Grun
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Group of Inflammation and Cellular Senescence and Laboratory of Molecular Biology and Bioinformatics, Department of Biochemistry/ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Nevton da Rosa Teixeira
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Group of Inflammation and Cellular Senescence and Laboratory of Molecular Biology and Bioinformatics, Department of Biochemistry/ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Lúcia von Mengden
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratory of Cellular Biochemistry, Department of Biochemistry/ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Marco Antônio de Bastiani
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratory of Cellular Biochemistry, Department of Biochemistry/ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Mariana Migliorini Parisi
- Interdisciplinary Group of Health (GIS), Center of Education and Research of the Institute of Cardiology of Cruz Alta, University of Cruz Alta (UNICRUZ), Cruz Alta, Brazil
| | - Rafael Bortolin
- Department of Civil and Environmental Engineering, Universidad de la Costa, Barranquilla, Atlántico, Colombia
| | - Patrícia Lavandoski
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Group of Inflammation and Cellular Senescence and Laboratory of Molecular Biology and Bioinformatics, Department of Biochemistry/ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Vinícius Pierdoná
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Group of Inflammation and Cellular Senescence and Laboratory of Molecular Biology and Bioinformatics, Department of Biochemistry/ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Letícia Biscaino Alves
- Center of Obesity and Metabolic Syndrome, São Lucas Hospital at Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS) and Postgraduate Program: Medicine and Health Sciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - José Cláudio Fonseca Moreira
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratory of Cellular Stress, Department of Biochemistry/ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Cláudio Corá Mottin
- Center of Obesity and Metabolic Syndrome, São Lucas Hospital at Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS) and Postgraduate Program: Medicine and Health Sciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil; Laboratory of Respiratory Physiology, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), School of Medicine, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Marcus Herbert Jones
- Laboratory of Respiratory Physiology, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), School of Medicine, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Fábio Klamt
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratory of Cellular Biochemistry, Department of Biochemistry/ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Alexandre Vontobel Padoin
- Center of Obesity and Metabolic Syndrome, São Lucas Hospital at Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS) and Postgraduate Program: Medicine and Health Sciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Fátima Costa Rodrigues Guma
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Group of Inflammation and Cellular Senescence and Laboratory of Molecular Biology and Bioinformatics, Department of Biochemistry/ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratory of Biochemistry and Cellular Biology of Lipids, Department of Biochemistry/ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Florencia María Barbé-Tuana
- Postgraduate Program: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Group of Inflammation and Cellular Senescence and Laboratory of Molecular Biology and Bioinformatics, Department of Biochemistry/ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Postgraduate Program in Cellular and Molecular Biology, School of Sciences, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil.
| |
Collapse
|
43
|
Ghosh Dastidar S, Jagatheesan G, Haberzettl P, Shah J, Hill BG, Bhatnagar A, Conklin DJ. Glutathione S-transferase P deficiency induces glucose intolerance via JNK-dependent enhancement of hepatic gluconeogenesis. Am J Physiol Endocrinol Metab 2018; 315:E1005-E1018. [PMID: 30153066 PMCID: PMC6293160 DOI: 10.1152/ajpendo.00345.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Hepatic glutathione S-transferases (GSTs) are dysregulated in human obesity, nonalcoholic fatty liver disease, and diabetes. The multifunctional GST pi-isoform (GSTP) catalyzes the conjugation of glutathione with acrolein and inhibits c-Jun NH2-terminal kinase (JNK) activation. Herein, we tested whether GSTP deficiency disturbs glucose homeostasis in mice. Hepatic GST proteins were downregulated by short-term high-fat diet in wild-type (WT) mice concomitant with increased glucose intolerance, JNK activation, and cytokine mRNAs in the liver. Genetic deletion of GSTP did not affect body composition, fasting blood glucose levels, or insulin levels in mice maintained on a normal chow diet; however, compared with WT mice, the GSTP-null mice were glucose intolerant. In GSTP-null mice, pyruvate intolerance, reflecting increased hepatic gluconeogenesis, was accompanied by elevated levels of activated JNK, cytokine mRNAs, and glucose-6-phosphatase proteins in the liver. Treatment of GSTP-null mice with the JNK inhibitor 1,9-pyrazoloanthrone (SP600125) significantly attenuated pyruvate-induced hepatic gluconeogenesis and significantly altered correlations between hepatic cytokine mRNAs and metabolic outcomes in GSTP-null mice. Collectively, these findings suggest that hepatic GSTP plays a pivotal role in glucose handling by regulating JNK-dependent control of hepatic gluconeogenesis. Thus, hepatic GSTP-JNK dysregulation may be a target of new therapeutic interventions during early stages of glucose intolerance to prevent the worsening metabolic derangements associated with human obesity and its relentless progression to diabetes.
Collapse
Affiliation(s)
- Shubha Ghosh Dastidar
- Diabetes and Obesity Center, School of Medicine, University of Louisville , Louisville, Kentucky
| | - Ganapathy Jagatheesan
- Diabetes and Obesity Center, School of Medicine, University of Louisville , Louisville, Kentucky
| | - Petra Haberzettl
- Diabetes and Obesity Center, School of Medicine, University of Louisville , Louisville, Kentucky
| | - Jasmit Shah
- Diabetes and Obesity Center, School of Medicine, University of Louisville , Louisville, Kentucky
- Department of Internal Medicine, Aga Khan University , Nairobi , Kenya
| | - Bradford G Hill
- Diabetes and Obesity Center, School of Medicine, University of Louisville , Louisville, Kentucky
| | - Aruni Bhatnagar
- Diabetes and Obesity Center, School of Medicine, University of Louisville , Louisville, Kentucky
| | - Daniel J Conklin
- Diabetes and Obesity Center, School of Medicine, University of Louisville , Louisville, Kentucky
| |
Collapse
|
44
|
Anderson EJ, Vistoli G, Katunga LA, Funai K, Regazzoni L, Monroe TB, Gilardoni E, Cannizzaro L, Colzani M, De Maddis D, Rossoni G, Canevotti R, Gagliardi S, Carini M, Aldini G. A carnosine analog mitigates metabolic disorders of obesity by reducing carbonyl stress. J Clin Invest 2018; 128:5280-5293. [PMID: 30226473 DOI: 10.1172/jci94307] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 09/11/2018] [Indexed: 12/15/2022] Open
Abstract
Sugar- and lipid-derived aldehydes are reactive carbonyl species (RCS) frequently used as surrogate markers of oxidative stress in obesity. A pathogenic role for RCS in metabolic diseases of obesity remains controversial, however, partly because of their highly diffuse and broad reactivity and the lack of specific RCS-scavenging therapies. Naturally occurring histidine dipeptides (e.g., anserine and carnosine) show RCS reactivity, but their therapeutic potential in humans is limited by serum carnosinases. Here, we present the rational design, characterization, and pharmacological evaluation of carnosinol, i.e., (2S)-2-(3-amino propanoylamino)-3-(1H-imidazol-5-yl)propanol, a derivative of carnosine with high oral bioavailability that is resistant to carnosinases. Carnosinol displayed a suitable ADMET (absorption, distribution, metabolism, excretion, and toxicity) profile and was determined to have the greatest potency and selectivity toward α,β-unsaturated aldehydes (e.g., 4-hydroxynonenal, HNE, ACR) among all others reported thus far. In rodent models of diet-induced obesity and metabolic syndrome, carnosinol dose-dependently attenuated HNE adduct formation in liver and skeletal muscle, while simultaneously mitigating inflammation, dyslipidemia, insulin resistance, and steatohepatitis. These improvements in metabolic parameters with carnosinol were not due to changes in energy expenditure, physical activity, adiposity, or body weight. Collectively, our findings illustrate a pathogenic role for RCS in obesity-related metabolic disorders and provide validation for a promising new class of carbonyl-scavenging therapeutic compounds rationally derived from carnosine.
Collapse
Affiliation(s)
- Ethan J Anderson
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, USA.,Department of Pharmacology and Toxicology, East Carolina University, Greenville, North Carolina, USA
| | - Giulio Vistoli
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Lalage A Katunga
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, North Carolina, USA
| | - Katsuhiko Funai
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA
| | - Luca Regazzoni
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - T Blake Monroe
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, USA.,Department of Pharmacology and Toxicology, East Carolina University, Greenville, North Carolina, USA
| | - Ettore Gilardoni
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Luca Cannizzaro
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Mara Colzani
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Danilo De Maddis
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Giuseppe Rossoni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | | | | | - Marina Carini
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
45
|
Abstract
The concept of cell signaling in the context of nonenzyme-assisted protein modifications by reactive electrophilic and oxidative species, broadly known as redox signaling, is a uniquely complex topic that has been approached from numerous different and multidisciplinary angles. Our Review reflects on five aspects critical for understanding how nature harnesses these noncanonical post-translational modifications to coordinate distinct cellular activities: (1) specific players and their generation, (2) physicochemical properties, (3) mechanisms of action, (4) methods of interrogation, and (5) functional roles in health and disease. Emphasis is primarily placed on the latest progress in the field, but several aspects of classical work likely forgotten/lost are also recollected. For researchers with interests in getting into the field, our Review is anticipated to function as a primer. For the expert, we aim to stimulate thought and discussion about fundamentals of redox signaling mechanisms and nuances of specificity/selectivity and timing in this sophisticated yet fascinating arena at the crossroads of chemistry and biology.
Collapse
Affiliation(s)
- Saba Parvez
- Department of Pharmacology and Toxicology, College of
Pharmacy, University of Utah, Salt Lake City, Utah, 84112, USA
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Marcus J. C. Long
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Jesse R. Poganik
- Ecole Polytechnique Fédérale de Lausanne,
Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Yimon Aye
- Ecole Polytechnique Fédérale de Lausanne,
Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
- Department of Biochemistry, Weill Cornell Medicine, New
York, New York, 10065, USA
| |
Collapse
|
46
|
Hauck AK, Zhou T, Hahn W, Petegrosso R, Kuang R, Chen Y, Bernlohr DA. Obesity-induced protein carbonylation in murine adipose tissue regulates the DNA-binding domain of nuclear zinc finger proteins. J Biol Chem 2018; 293:13464-13476. [PMID: 30012885 DOI: 10.1074/jbc.ra118.003469] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/20/2018] [Indexed: 11/06/2022] Open
Abstract
In obesity-linked insulin resistance, oxidative stress in adipocytes leads to lipid peroxidation and subsequent carbonylation of proteins by diffusible lipid electrophiles. Reduction in oxidative stress attenuates protein carbonylation and insulin resistance, suggesting that lipid modification of proteins may play a role in metabolic disease, but the mechanisms remain incompletely understood. Herein, we show that in vivo, diet-induced obesity in mice surprisingly results in preferential carbonylation of nuclear proteins by 4-hydroxy-trans-2,3-nonenal (4-HNE) or 4-hydroxy-trans-2,3-hexenal (4-HHE). Proteomic and structural analyses revealed that residues in or around the sites of zinc coordination of zinc finger proteins, such as those containing the C2H2 or MATRIN, RING, C3H1, or N4-type DNA-binding domains, are particularly susceptible to carbonylation by lipid aldehydes. These observations strongly suggest that carbonylation functionally disrupts protein secondary structure supported by metal coordination. Analysis of one such target, the nuclear protein estrogen-related receptor γ (ERR-γ), showed that ERR-γ is modified by 4-HHE in the obese state. In vitro carbonylation decreased the DNA-binding capacity of ERR-γ and correlated with the obesity-linked down-regulation of many key genes promoting mitochondrial bioenergetics. Taken together, these findings reveal a novel mechanistic connection between oxidative stress and metabolic dysfunction arising from carbonylation of nuclear zinc finger proteins, such as the transcriptional regulator ERR-γ.
Collapse
Affiliation(s)
- Amy K Hauck
- From the Departments of Biochemistry, Molecular Biology, and Biophysics and
| | - Tong Zhou
- From the Departments of Biochemistry, Molecular Biology, and Biophysics and
| | - Wendy Hahn
- From the Departments of Biochemistry, Molecular Biology, and Biophysics and
| | - Raphael Petegrosso
- Computer Science and Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455
| | - Rui Kuang
- Computer Science and Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455
| | - Yue Chen
- From the Departments of Biochemistry, Molecular Biology, and Biophysics and
| | - David A Bernlohr
- From the Departments of Biochemistry, Molecular Biology, and Biophysics and
| |
Collapse
|
47
|
Tranberg B, Hellgren LI, Lykkesfeldt J, Hansen AK. High-fat feeding induces mobilization of vitamin C in obese prone rats. Res Vet Sci 2018; 119:167-169. [PMID: 29945036 DOI: 10.1016/j.rvsc.2018.06.011] [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: 02/07/2018] [Revised: 06/06/2018] [Accepted: 06/10/2018] [Indexed: 11/30/2022]
Abstract
In obesity and dyslipidemia, hydrolysis of triacylglycerol (TAG) into non-esterified fatty acids (NEFAs) may contribute to insulin resistance, and production of oxygenated, bioactive polyunsaturated fatty acids may increase oxidative stress. Here we show that after six weeks of high-fat feeding of obese prone rats (Crl:OP(CD), vitamin C was increased both in liver (P < 0.01) and plasma (P < 0.001), while both TAG (P < 0.01) and NEFA (P < 0.001) were lower than in low-fat fed control rats. Hepatic vitamin C biosynthesis was similar between groups, indicating that a new steady state level was established with a higher vitamin C level adequate for supplying the systemic needs. Glucose and insulin sensitivity were unaffected at this stage. Eventually, the mobilization of vitamin C may be seen as a mechanism to protect the host against insulin resistance.
Collapse
Affiliation(s)
- B Tranberg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - L I Hellgren
- Department of Systems Biology, Center for Biological Sequence Analysis, Technical University of Denmark, Lyngby, Denmark
| | - J Lykkesfeldt
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - A K Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark.
| |
Collapse
|
48
|
A. R, Agrawal N, Kumar H, Kumar V. Emerging role of branched chain amino acids in metabolic disorders: A mechanistic review. PHARMANUTRITION 2018. [DOI: 10.1016/j.phanu.2018.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
49
|
Jia J, Liu Z, Xiao X, Liu B, Chou KC. iCar-PseCp: identify carbonylation sites in proteins by Monte Carlo sampling and incorporating sequence coupled effects into general PseAAC. Oncotarget 2018; 7:34558-70. [PMID: 27153555 PMCID: PMC5085176 DOI: 10.18632/oncotarget.9148] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 04/09/2016] [Indexed: 01/22/2023] Open
Abstract
Carbonylation is a posttranslational modification (PTM or PTLM), where a carbonyl group is added to lysine (K), proline (P), arginine (R), and threonine (T) residue of a protein molecule. Carbonylation plays an important role in orchestrating various biological processes but it is also associated with many diseases such as diabetes, chronic lung disease, Parkinson's disease, Alzheimer's disease, chronic renal failure, and sepsis. Therefore, from the angles of both basic research and drug development, we are facing a challenging problem: for an uncharacterized protein sequence containing many residues of K, P, R, or T, which ones can be carbonylated, and which ones cannot? To address this problem, we have developed a predictor called iCar-PseCp by incorporating the sequence-coupled information into the general pseudo amino acid composition, and balancing out skewed training dataset by Monte Carlo sampling to expand positive subset. Rigorous target cross-validations on a same set of carbonylation-known proteins indicated that the new predictor remarkably outperformed its existing counterparts. For the convenience of most experimental scientists, a user-friendly web-server for iCar-PseCp has been established at http://www.jci-bioinfo.cn/iCar-PseCp, by which users can easily obtain their desired results without the need to go through the complicated mathematical equations involved. It has not escaped our notice that the formulation and approach presented here can also be used to analyze many other problems in computational proteomics.
Collapse
Affiliation(s)
- Jianhua Jia
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen 333403 China.,Gordon Life Science Institute, Boston, MA 02478, USA
| | - Zi Liu
- School of Computer Science and Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Xuan Xiao
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen 333403 China.,Gordon Life Science Institute, Boston, MA 02478, USA
| | - Bingxiang Liu
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen 333403 China
| | - Kuo-Chen Chou
- Gordon Life Science Institute, Boston, MA 02478, USA.,Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
50
|
Dysregulation of mitochondrial function and biogenesis modulators in adipose tissue of obese children. Int J Obes (Lond) 2017; 42:618-624. [PMID: 29158541 DOI: 10.1038/ijo.2017.274] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 10/01/2017] [Accepted: 10/17/2017] [Indexed: 12/26/2022]
Abstract
BACKGROUND/OBJECTIVES We aimed to evaluate mitochondrial biogenesis (MB), structure, metabolism and dysfunction in abdominal adipose tissue from male pediatric patients with obesity. SUBJECTS/METHODS Samples were collected from five children with obesity (percentile ⩾95) and five eutrophic boys (percentile ⩾5/⩽85) (8-12 years old) following parental informed consent. We analyzed the expression of key genes involved in MB (sirtuin-1 (SIRT1), peroxisome proliferator-activated receptor-γ (PPARγ), PPARγ coactivator-1α (PGC1α), nuclear respiratory factors 1 and 2 (NRF1, NRF2) and mitochondrial transcription factor A (TFAM) and surrogates for mitochondrial function/structure/metabolism (porin, TOMM20, complex I and V, UCP1, UCP2, SIRT3, SOD2) by western blot. Citrate synthase (CS), complex I (CI) activity, adenosine triphosphate (ATP) levels, mitochondrial DNA (mtDNA) content and oxidative stress end points were also determined. RESULTS Most MB proteins were significantly decreased in samples from children with obesity except complex I, V and superoxide dismutase-2 (SOD2). Similarly, CS and CI activity showed a significant reduction, as well as ATP levels and mtDNA content. PPARγ, PGC1α, complex I and V and SOD2 were hyperacetylated compared with lean samples. Concurrently, in samples from children with obesity, we found decreased SOD2 activity and redox state imbalance highlighted by decreased reduced glutathione/oxidized glutathione (GSH/GSSG) ratio and significant increases in protein carbonylation. CONCLUSIONS Adipose tissue from children with obesity demonstrates a dysregulation of key modulators of MB and organelle structure, and displays hyperacetylation of key proteins and altered expression of upstream regulators of cell metabolism.
Collapse
|