1
|
Chai J, Xu J, Zhang S, Yan W, Chen S, Zhu X, Luo C, Wang W. CCAAT/Enhancer-Binding Protein Beta Nitration Participates in Hyperhomocysteinemia-Induced Cardiomyocyte Autophagic Flux Blockage by Inhibiting Transcription Factor EB Transcription. Antioxid Redox Signal 2025. [PMID: 39783266 DOI: 10.1089/ars.2023.0517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Aims: Autophagy is a protective mechanism of cardiomyocytes. Hyperhomocysteinemia (HHcy) elevates oxidative and nitrosative stress levels, leading to an abnormal increase in nitration protein, possibly leading to abnormal autophagy regulation in cardiomyocytes. However, the regulatory effect of HHcy on autophagy at the post-translational modification level is still unclear. Here, we aimed to explore the regulatory mechanism of HHcy on transcription factor EB (TFEB) and nitration of CCAAT/enhancer-binding protein beta (C/EBPβ), a transcriptional repressor of Tfeb, on autophagy in cardiomyocytes. Results: In this study, we established the HHcy rat model by feeding a 2.5% (w/w) methionine diet. The nitration level of C/EBPβ was increased in HHcy, which promoted the entry of C/EBPβ into the nucleus, enhanced the transcriptional suppressive effect of C/EBPβ on Tfeb, and induced insufficient autophagy in cardiomyocytes. Furthermore, we confirmed that the Tyr 274 site of C/EBPβ could undergo nitration induced by HHcy. Once C/EBPβ was nitrated on the Tyr 274 site, the nuclear translocation of C/EBPβ and transcription suppressor function of C/EBPβ on Tfeb were enhanced. Innovation and Conclusion: We find that C/EBPβ is a transcriptional repressor of Tfeb, and HHcy induces the nitration at the Tyr 274 site of C/EBPβ, leading to autophagic flux blockage in cardiomyocytes. These data indicated that nitrated C/EBPβ might be a potential therapeutic target against HHcy-induced autophagy insufficiency of cardiomyocytes. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Jiayin Chai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jiahui Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shangyue Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wenjing Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuai Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinyu Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chenghua Luo
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, China
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Zhang WW, Xiang Y, Chen L, Liu ST, Lin CC, Li JX, Xiang LX, Chen NX, Shi DL, Zhang YY, Wang XY, Hu LY, Chen S, Luo Y, Tan CN, Xue PP, Jiang YZ, Li SWC, Yang ZX, Dai JG, Li ZJ, Ran Q. Dietary methionine supplementation promotes mice hematopoiesis after irradiation. Mil Med Res 2024; 11:83. [PMID: 39702305 DOI: 10.1186/s40779-024-00584-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 11/16/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND With the increasing risk of nuclear exposure, more attention has been paid to the prevention and treatment of acute radiation syndrome (ARS). Although amino acids are key nutrients involved in hematopoietic regulation, the impacts of amino acids on bone marrow hematopoiesis following irradiation and the associated mechanisms have not been fully elucidated. Hence, it is of paramount importance to study the changes in amino acid metabolism after irradiation and their effects on hematopoiesis as well as the related mechanisms. METHODS The content of serum amino acids was analyzed using metabolomic sequencing. The survival rate and body weight of the irradiated mice were detected after altering the methionine content in the diet. Extracellular matrix (ECM) protein analysis was performed via proteomics analysis. Inflammatory factors were examined by enzyme-linked immunosorbent assay (ELISA). Flow cytometry, Western blotting, and immunofluorescence were employed to determine the mechanism by which S100 calcium-binding protein A4 (S100A4) regulates macrophage polarization. RESULTS The survival time of irradiated mice was significantly associated with alterations in multiple amino acids, particularly methionine. A high methionine diet promoted irradiation tolerance, especially in the recovery of bone marrow hematopoiesis, yet with dose limitations. Folate metabolism could partially alleviate the dose bottleneck by reducing the accumulation of homocysteine. Mechanistically, high methionine levels maintained the abundance of ECM components, including collagens and glycoproteins, in the bone marrow post-irradiation, among which the level of S100A4 was significantly changed. S100A4 regulated macrophage polarization via the STAT3 pathway, inhibited bone marrow inflammation and facilitated the proliferation and differentiation of hematopoietic stem/progenitor cells. CONCLUSIONS We have demonstrated that an appropriate elevation in dietary methionine enhances irradiation tolerance in mice and explains the mechanism by which methionine regulates bone marrow hematopoiesis after irradiation.
Collapse
Affiliation(s)
- Wei-Wei Zhang
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Yang Xiang
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Li Chen
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Shao-Ting Liu
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
- Department of Nutrition, The Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
| | - Chuan-Chuan Lin
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Jiu-Xuan Li
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Li-Xin Xiang
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Nan-Xi Chen
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Dong-Ling Shi
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Yang-Yang Zhang
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Xue-Ying Wang
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Lan-Yue Hu
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Sai Chen
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Ya Luo
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Cheng-Ning Tan
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Pei-Pei Xue
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Yang-Zhou Jiang
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China
| | - Sheng-Wen Calvin Li
- Neuro-Oncology and Stem Cell Research Laboratory, CHOC Children's Research Institute, Children's Hospital of Orange County (CHOC), Orange, CA, 92868-3874, USA
- Department of Neurology, University of California - Irvine School of Medicine, Orange, CA, 92868, USA
| | - Zhen-Xing Yang
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China.
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China.
| | - Ji-Gang Dai
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China.
- Department of Thoracic Surgery, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China.
| | - Zhong-Jun Li
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China.
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China.
| | - Qian Ran
- Department of Blood Transfusion, Laboratory Medicine Center, the Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China.
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing, 400037, China.
| |
Collapse
|
3
|
India-Aldana S, Midya V, Betanzos-Robledo L, Yao M, Alcalá C, Andra SS, Arora M, Calafat AM, Chu J, Deierlein A, Estrada-Gutierrez G, Jagani R, Just AC, Kloog I, Landero J, Oulhote Y, Walker RW, Yelamanchili S, Baccarelli AA, Wright RO, Téllez Rojo MM, Colicino E, Cantoral A, Valvi D. Impact of metabolism-disrupting chemicals and folic acid supplementation on liver injury and steatosis in mother-child pairs. J Hepatol 2024:S0168-8278(24)02757-0. [PMID: 39674324 DOI: 10.1016/j.jhep.2024.11.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND AND AIMS Scarce knowledge about the impact of metabolism-disrupting chemicals (MDCs) on steatotic liver disease limits opportunities for intervention. We evaluated pregnancy MDC-mixture associations with liver outcomes, and effect modification by folic acid (FA) supplementation in mother-child pairs. METHODS We studied ∼200 mother-child pairs from the Mexican PROGRESS cohort, with measured 43 MDCs during pregnancy (estimated air pollutants, blood/urine metals or metalloids, urine high- and low-molecular-weight phthalate [HMWPs, LMWPs] and organophosphate-pesticide [OP] metabolites), and serum liver enzymes (ALT, AST) at ∼9 years post-parturition. Outcomes included elevated liver enzymes in children and established clinical scores for steatosis and fibrosis in mothers (i.e. , AST ALT, FLI, HSI, FIB-4). Bayesian Weighted Quantile Sum regression assessed MDC-mixture associations with liver outcomes. We further examined chemical-chemical interactions and effect modification by self-reported FA supplementation. RESULTS In children, many MDC-mixtures were associated with liver injury. Per quartile HMWP-mixture increase, ALT increased by 10.1% (95%CI: 1.67%, 19.4%) and AST by 5.27% (95% CI: 0.80%, 10.1%). LMWP-mixtures and air pollutant-mixtures were associated with higher AST and ALT, respectively. Air pollutant and non-essential metal/element associations with liver enzymes were attenuated by maternal cobalt blood concentrations (p-interactions<0.05). In mothers, only the LMWP-mixture was associated with odds for steatosis [OR=1.53 (95%CI: 1.01, 2.28) for HSI>36, and OR=1.62 (95%CI: 1.05, 2.49) for AST:ALT<1]. In mothers and children, most associations were attenuated (null) at FA supplementation≥600mcg/day (p-interactions<0.05). CONCLUSIONS Pregnancy MDC exposures may increase risk for liver injury and steatosis, particularly in children. Adequate FA supplementation and maternal cobalt levels may attenuate these associations. IMPACT AND IMPLICATIONS The effects of environmental chemical exposures on steatotic liver diseases are not well understood. In a parallel investigation of mothers and children, we found that pregnancy exposures to metabolism-disrupting chemicals may increase the risk for liver injury and steatosis, especially in the child, and that these associations could be attenuated by higher folic acid and/or cobalt levels. These findings can inform policies to decrease environmental chemical pollution and contribute to the design of clinical interventions addressing the MASLD epidemic.
Collapse
Affiliation(s)
- Sandra India-Aldana
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vishal Midya
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Larissa Betanzos-Robledo
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Meizhen Yao
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cecilia Alcalá
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Syam S Andra
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manish Arora
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Antonia M Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Jaime Chu
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea Deierlein
- New York University School of Global Public Health, New York, NY, USA
| | | | - Ravikumar Jagani
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Allan C Just
- Department of Epidemiology, School of Public Health, Brown University, Providence, RI, USA
| | - Itai Kloog
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julio Landero
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Youssef Oulhote
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ryan W Walker
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shirisha Yelamanchili
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Robert O Wright
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martha María Téllez Rojo
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Elena Colicino
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Damaskini Valvi
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Zhu J, Liao X, Du L, Lv P, Deng J. Associations of serum folate and vitamin B 12 levels with all-cause mortality among patients with metabolic dysfunction associated steatotic liver disease: a prospective cohort study. Front Endocrinol (Lausanne) 2024; 15:1426103. [PMID: 39703860 PMCID: PMC11655224 DOI: 10.3389/fendo.2024.1426103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 11/11/2024] [Indexed: 12/21/2024] Open
Abstract
Introduction Serum folate and vitamin B12 levels correlate with the prevalence of fatty liver disease, but it is not clear how they affect mortality. Therefore, this study aimed to investigate the association of serum folate and vitamin B12 concentrations with all-cause mortality in individuals with metabolic dysfunction-associated steatotic liver disease (MASLD). Methods MASLD subjects were from the Third National Health and Nutrition Examination Survey (NHANES III) in the United States, and mortality follow-up data were obtained by linkage to death records from the National Death Index. Multivariable Cox proportional regression models and restricted cubic spline (RCS) models were used to evaluate the association of serum folate/vitamin B12 with all-cause mortality in the MASLD population. Results 3,636 and 2,125 MASLD individuals were included in the analyses related to serum folate and vitamin B12, respectively. During a follow-up period of more than 20 years, the RCS models demonstrated significant nonlinear associations of both serum folate (P <0.001) and vitamin B12 (P =0.016) with all-cause mortality in MASLD. When their serum concentrations were below the median level, the risk of all-cause mortality decreased with increasing concentration, reaching a lowest risk around the median level, and then leveled off. In the multivariable cox regression model, for vitamin B12, the risk of all-cause mortality was reduced by 42% and 28% in the third and fourth quartile groups, respectively, compared with the lowest quartile group (hazard ratio [HR]=0.58, 95% CI: 0.39-0.86, P =0.008; HR =0.72, 95% CI: 0.54-0.96, P=0.026, respectively). For folate, the risk of all-cause mortality was reduced by 28% in the third quartile compared with the lowest quartile (HR =0.72, 95% CI: 0.57-0.91, P =0.005). Conclusion This longitudinal cohort study suggests that low serum folate and vitamin B12 levels in patients with MASLD are significantly associated with an elevated risk of all-cause mortality.
Collapse
Affiliation(s)
- Jiaxin Zhu
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xinyi Liao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Du
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Pengju Lv
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jian Deng
- Department of Thyroid Breast Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
5
|
Abdi F, Farhangi MA, Mohammadzadeh M. Habitual dietary methyl donor's intake and metabolic profile in obese individuals: a cross-sectional study. Sci Rep 2024; 14:30046. [PMID: 39627237 PMCID: PMC11615318 DOI: 10.1038/s41598-024-75388-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 10/04/2024] [Indexed: 12/06/2024] Open
Abstract
Considering the role of dietary methyl donor (DMD) in numerous biochemical processes, we hypothesized that DMD could play an important role in metabolic syndrome such as hyperlipidemia, hypertension, insulin resistance, and appetite in obese individuals. This cross-sectional study was conducted on 335 obese people. We collected dietary data using a valid and reliable 147-question Food Frequency Questionnaire (FFQ). Multivariate multinomial logistic regression was used to estimate the odds ratio (OR) and 95% confidence interval (CI) for the association between dietary methyl intake and cardio-metabolic risk factors. After adjusting for confounding variables, individuals at the fourth and third quartile of DMD, were more likely to have lower low-density lipoprotein cholesterol (LDL-C) (OR = 0.968, CI = 0.943-0.994, P = 0.015 and OR = 0.978, CI = 0.957-0.998, P = 0.03 respectively) versus first quartile. Also, total cholesterol (TC) showed a significant decrease in forth quartile of DMD in model III (OR = 0.974, CI = 0.951-0.997, P = 0.029). Current results suggested that, high DMDs' consumption, significantly associated with decreased risk of cardiometabolic risk factors.
Collapse
Affiliation(s)
- Fatemeh Abdi
- Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Milad Mohammadzadeh
- Department of Clinical Nutrition and Dietetic, School of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Dong J, Li Z, Wang C, Zhang R, Li Y, Liu M, Chen Q, Bai Y, Lv W. Dietary folate intake and all-cause mortality and cardiovascular mortality in American adults with non-alcoholic fatty liver disease: Data from NHANES 2003 to 2018. PLoS One 2024; 19:e0314148. [PMID: 39570932 PMCID: PMC11581259 DOI: 10.1371/journal.pone.0314148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/05/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND The relationship between dietary folate intake and prior mortality in adult patients with Non-alcoholic Fatty Liver Disease (NAFLD) has not been clearly studied. We aimed to examine the relationship between dietary folate intake and all-cause and cardiovascular (CVD) mortality in adult NAFLD patients in the US. METHODS Using data from National Health and Nutrition Examination Survey (NHANES) 2003-2018 and associated mortality data we conducted a cohort study of US adult NAFLD subjects. Multivariable Cox proportional hazards regression models were used to evaluate the relationship between dietary folate intake and both all-cause mortality and CVD mortality, accounting for potential confounders. The study employed restricted cubic spline analysis to investigate the non-linear association between dietary folate levels and mortality from all causes and cardiovascular disease. RESULTS Our final cohort consisted of 3,266 NAFLD patients, with a median follow-up of 10.3 years, 691 deaths were observed, including 221 cardiovascular deaths. Compared to participants with a folate intake in Quartile 1 (≤250 μg/d), those in Quartile 4 (≥467.5 μg/d) had multivariable-adjusted hazard ratios of 0.69 (95% CI, 0.51-0.94) for all-cause mortality (p for trend = 0.028) and 0.55 (95% CI, 0.29-1.04) for CVD mortality (p for trend = 0.107). A non-linear relationship between dietary intake and risk of death was not observed. CONCLUSION Greater dietary folate intake is associated with a reduced risk of all-cause in American adults with NAFLD. Higher dietary folate intake not found to be associated with lower CVD mortality. These findings suggest that dietary folate may improve the prognosis of adult NAFLD patients. The measured-response relationship between dietary folate intake and mortality in patients with NAFLD requires further investigation.
Collapse
Affiliation(s)
- Jinsheng Dong
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiqiang Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Chenlu Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Runshun Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yilin Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mingkun Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiuye Chen
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuning Bai
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenliang Lv
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Zhu X, Cao Y, Chen S, Liu Q, Chai J, Wang W. Insufficient S-sulfhydration of serum and glucocorticoid-regulated kinase 1 participates in hyperhomocysteinemia-induced liver injury. Free Radic Biol Med 2024; 225:517-527. [PMID: 39427745 DOI: 10.1016/j.freeradbiomed.2024.10.294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/01/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND & AIMS Previous studies have established that hyperhomocysteinemia (HHcy) significantly contributes to the development of non-alcoholic steatohepatitis (NASH). Conversely, hydrogen sulfide (H2S) has shown potential in mitigating NASH. Despite these findings, it remains uncertain whether H2S can serve as a therapeutic agent against HHcy-induced liver damage. METHODS Mice were fed a high-methionine diet to induce HHcy and HepG2 cells were exposed to homocysteine (Hcy). In both models, we assessed liver injury, H2S concentration, and autophagy levels. For rescue, sodium hydrosulfide (NaHS), an H2S donor, was used to test its potential in reversing hepatic pathological features induced by HHcy. RESULTS 1) Hcy accumulation led to liver damage and increased autophagy. This was linked to insufficient S-sulfhydration of serum and glucocorticoid-regulated kinase 1 (SGK1) at Cys244 and Cys282, a crucial autophagy regulator. The deficiency in S-sulfhydration was resulted from downregulation of cystathionine-γ-lyase (CSE) and subsequent H2S decrease, leading to SGK1 inactivation. 2) Administration of NaHS reduced the liver damage caused by high Hcy levels and restored H2S levels, promoting the S-sulfhydration and activation of SGK1. 3) Pharmacological inhibition of SGK1 induced autosis, a specific type of cell death caused by overactivation of autophagy. Conversely, a constitutively active mutant of SGK1 (SGK1S422D) significantly decreased autophagy and improved cell viability. CONCLUSIONS NaHS supplementation mitigates HHcy-induced liver injury by downregulating hepatic autophagy through the S-sulfhydration and activation of SGK1. This post-translational modification by H2S holds promise as a therapeutic approach for HHcy-induced liver injury.
Collapse
Affiliation(s)
- Xinyu Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yan Cao
- Department of Basic Medical Sciences, Beijing Health Vocational College, Beijing, 101149, China
| | - Shuai Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Qinchi Liu
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jiayin Chai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, 100069, China.
| |
Collapse
|
8
|
Wang L, Gao J, Ma J, Sun J, Wang Y, Luo J, Wang Z, Wang H, Li J, Yang D, Wang J, Hu R. Effects of hyperhomocysteinemia on follicular development and oocytes quality. iScience 2024; 27:111241. [PMID: 39563894 PMCID: PMC11574796 DOI: 10.1016/j.isci.2024.111241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/12/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
In patients with polycystic ovary syndrome (PCOS), the concentration of homocysteine (Hcy) in follicular fluid is inversely correlated with oocyte and embryo quality. Nevertheless, other metabolic abnormalities associated with PCOS may also impact oocyte and early embryo quality. Therefore, it remains uncertain whether reproductive function is affected in patients without PCOS with hyperhomocysteinemia (HHcy). Here, we observed reduced fertility, increased ovarian atretic follicles, and reduced oocyte maturation rates in HHcy mice. Proteomic analyses revealed that HHcy causes mitochondrial dysfunction and reduced expression of zona pellucida proteins (ZP1, ZP2, and ZP3) in oocytes. Transmission electron microscopy confirmed abnormal formation of the zona pellucida and microvilli in oocytes from HHcy mice. Additionally, in vitro fertilization (IVF) demonstrated a reduction in the rate of 2-cell embryo formation in HHcy mice. These findings reveal that HHcy reduces female reproductive longevity by affecting follicular development and oocyte quality.
Collapse
Affiliation(s)
- Lu Wang
- Ningxia Medical University, General Hospital of Ningxia Medical University, Ningxia, China
| | - Jinmei Gao
- Ningxia Medical University, General Hospital of Ningxia Medical University, Ningxia, China
| | - Jie Ma
- Ningxia Medical University, General Hospital of Ningxia Medical University, Ningxia, China
| | - Jing Sun
- Ningxia Medical University, Ningxia, China
| | - Yajie Wang
- Ningxia Medical University, General Hospital of Ningxia Medical University, Ningxia, China
| | - Jia Luo
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Ningxia, China
| | | | - Hui Wang
- Reproductive Medicine Center, Yinchuan Women and Children Healthcare Hospital, Ningxia, China
| | - Jialing Li
- Reproductive Medicine Center, General Hospital of Ningxia Medical University, Ningxia, China
| | - Danyu Yang
- Ningxia Medical University, General Hospital of Ningxia Medical University, Ningxia, China
| | - Jinfang Wang
- Department of Obstetrician, General Hospital of Ningxia Medical University, Ningxia, China
| | - Rong Hu
- Reproductive Medicine Center, General Hospital of Ningxia Medical University, Ningxia, China
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Ningxia, China
| |
Collapse
|
9
|
Chen Y, Li X, Lu R, Lv Y, Wu Y, Ye J, Zhao J, Li L, Huang Q, Meng W, Long F, Huang W, Xia Q, Yu J, Fan C, Mo X. Vitamin B 12 protects necrosis of acinar cells in pancreatic tissues with acute pancreatitis. MedComm (Beijing) 2024; 5:e686. [PMID: 39415850 PMCID: PMC11480517 DOI: 10.1002/mco2.686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/23/2024] [Accepted: 06/28/2024] [Indexed: 10/19/2024] Open
Abstract
Pharmacological agents regarding the most optimal treatments of acute pancreatitis remain. One-carbon metabolism nutrients as therapeutic agents in many diseases might be involved in acute pancreatitis. The roles are acquired exploration in acute pancreatitis. We utilized Mendelian randomization to assess the causal impact of folate, homocysteine, and vitamin B12 (VB12) on acute pancreatitis. Wild-type and corresponding genetically modified mouse models were used to verify the genetic correlating findings. A negative association between genetically predicted serum VB12 levels and risks of acute pancreatitis was identified in human population. The transcobalamin receptor (TCblR)/CD320 gene ablation that decreased cellular VB12 uptake and ATP production in pancreatic tissues promoted necrosis, resulting in much severe pathological changes of induced acute pancreatitis in mice. VB12 pretreatment and posttreatment dramatically increased ATP levels in pancreatic tissues and reduced the necrosis, then the elevated levels of amylase in serum, the levels of CK-19, the activity of trypsin, and T lymphocyte infiltration in pancreatic tissues, prevented the pancreatic gross loss and ameliorated histopathological changes of mouse pancreases with induced acute pancreatitis. The results reveal that VB12 is potential as a therapeutic agent to inhibit tissue injuries and adaptive inflammatory responses in the pancreas in patients with acute pancreatitis.
Collapse
Affiliation(s)
- Yulin Chen
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineLaboratory of Stem Cell BiologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xue Li
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineLaboratory of Stem Cell BiologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Ran Lu
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineLaboratory of Stem Cell BiologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
- Department of Occupational and Environmental HealthWest China School of Public Health and West China Fourth HospitalSichuan UniversityChengduChina
- West China‐PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan UniversityChengduChina
| | - Yinchun Lv
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineLaboratory of Stem Cell BiologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Yongzi Wu
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineLaboratory of Stem Cell BiologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Junman Ye
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineLaboratory of Stem Cell BiologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Jin Zhao
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineLaboratory of Stem Cell BiologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Li Li
- School of Basic MedicineSouthwest Medical UniversityLuzhouChina
| | - Qiaorong Huang
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineLaboratory of Stem Cell BiologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Wentong Meng
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineLaboratory of Stem Cell BiologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Feiwu Long
- Department of GastrointestinalBariatric, and Metabolic SurgeryResearch Center for NutritionMetabolism & Food SafetyWest China‐PUMC C.C. Chen Institute of HealthWest China School of Public Health and West China Fourth Hospital, Sichuan UniversityChengduChina
| | - Wei Huang
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineLaboratory of Stem Cell BiologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Qing Xia
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineLaboratory of Stem Cell BiologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Jianbo Yu
- Longgang Central HospitalShenzhenChina
| | - Chuanwen Fan
- Department of GastrointestinalBariatric, and Metabolic SurgeryResearch Center for NutritionMetabolism & Food SafetyWest China‐PUMC C.C. Chen Institute of HealthWest China School of Public Health and West China Fourth Hospital, Sichuan UniversityChengduChina
- Department of Oncology and Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden
| | - Xianming Mo
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineLaboratory of Stem Cell BiologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
10
|
Kurata H, Meguro S, Abe Y, Sasaki T, Arai Y, Hayashi K. Association of fish intake with all-cause mortality according to CRP levels or inflammation in older adults: a prospective cohort study. BMC Public Health 2024; 24:2822. [PMID: 39407192 PMCID: PMC11481737 DOI: 10.1186/s12889-024-20162-z] [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: 03/08/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND The relationship between inflammatory response, fish consumption, and mortality risk in older individuals is unclear. We investigated whether C-reactive protein (CRP) levels ≥ 0.1 mg/dL, fish intake, and inflammatory responses are associated with all-cause mortality risk in older adults. METHODS This prospective cohort study included older adults aged 85-89 years from the Kawasaki Aging and Wellbeing Project, who did not require daily care. Cohort was recruited from March 2017 to December 2018 (follow-up ended on December 31, 2021). Dietary assessment was conducted using the Brief Self-Administered Diet History Questionnaire. Multivariate Cox proportional hazards regression was used to estimate the hazard ratio (HR) and 95% confidence interval (CI) for all-cause mortality in the CRP ≥ 0.1 mg/dL group; the CRP < 0.1 mg/dL group was used for reference. Within CRP ≥ 0.1 and < 0.1 mg/dL groups, participants were categorized into tertiles of fish intake. HRs and 95% CIs for all-cause mortality in the other groups were estimated using the lower tertile group as a reference. RESULTS The study included 996 participants (mean [standard deviation] age, 86.5 [1.37] years; 497 [49.9%] women) with a median CRP level of 0.08 (interquartile range [IQR] = 0.04-0.16). There were 162 deaths during 4,161 person-years of observation; the multivariable-adjusted HR for all-cause mortality in the CRP ≥ 0.1 mg/dL group was 1.86 (95% CI, 1.32-2.62); P < 0.001. In 577 individuals with median (IQR) fish intake of 39.3 g/1000 kcal (23.6-57.6) and CRP level of < 0.1 mg/dL, the multivariable-adjusted HR for all-cause mortality in the higher tertile group of fish intake was 1.15 (0.67-1.97); P = 0.59, non-linear P = 0.84. In 419 individuals with median (IQR) fish intake of 40.7 g/1000 kcal (25.0-60.1) and CRP level of ≥ 0.1 mg/dL, the multivariate-adjusted HR for all-cause mortality in the higher tertile group of fish intake was 0.49 (0.26-0.92); P = 0.026, non-linear P = 0.38, P-value for interaction = 0.040. CONCLUSIONS A negative association between fish intake and all-cause mortality was seen in older adults with elevated CRP levels, which is a mortality risk factor. While the results may be limited owing to stringent methods ensuring impartiality, they offer valuable insights for future research. TRIAL REGISTRATION UMIN000026053. Registered February 24, 2017.
Collapse
Affiliation(s)
- Hideaki Kurata
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Shu Meguro
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, 160-0016, Japan.
| | - Yukiko Abe
- Centre for Supercentenarian Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Takashi Sasaki
- Centre for Supercentenarian Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Yasumichi Arai
- Centre for Supercentenarian Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Kaori Hayashi
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, 160-0016, Japan
| |
Collapse
|
11
|
Rho H, Kim S, Kim SU, Kim JW, Lee SH, Park SH, Escorcia FE, Chung JY, Song J. CHIP ameliorates nonalcoholic fatty liver disease via promoting K63- and K27-linked STX17 ubiquitination to facilitate autophagosome-lysosome fusion. Nat Commun 2024; 15:8519. [PMID: 39353976 PMCID: PMC11445385 DOI: 10.1038/s41467-024-53002-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 09/26/2024] [Indexed: 10/03/2024] Open
Abstract
The fusion of autophagosomes and lysosomes is essential for the prevention of nonalcoholic fatty liver disease (NAFLD). Here, we generate a hepatocyte-specific CHIP knockout (H-KO) mouse model that develops NAFLD more rapidly in response to a high-fat diet (HFD) or high-fat, high-fructose diet (HFHFD). The accumulation of P62 and LC3 in the livers of H-KO mice and CHIP-depleted cells indicates the inhibition of autophagosome-lysosome fusion. AAV8-mediated overexpression of CHIP in the murine liver slows the progression of NAFLD induced by HFD or HFHFD feeding. Mechanistically, CHIP induced K63- and K27-linked polyubiquitination at the lysine 198 residue of STX17, resulting in increased STX17-SNAP29-VAMP8 complex formation. The STX17 K198R mutant was not ubiquitinated by CHIP; it interfered with its interaction with VAMP8, rendering STX17 incapable of inhibiting steatosis development in mice. These results indicate that a signaling regulatory mechanism involving CHIP-mediated non-degradative ubiquitination of STX17 is necessary for autophagosome-lysosome fusion.
Collapse
Affiliation(s)
- Hyunjin Rho
- Department of Biochemistry, College of Life Science and Technology, Institute for Bio-medical Convergence Science and Technology, Yonsei University, Seoul, Republic of Korea
| | - Seungyeon Kim
- Department of Biochemistry, College of Life Science and Technology, Institute for Bio-medical Convergence Science and Technology, Yonsei University, Seoul, Republic of Korea
| | - Seung Up Kim
- Department of Internal Medicine, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Jeong Won Kim
- Department of Pathology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Sang Hoon Lee
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
- GENINUS Inc, Seoul, Republic of Korea
| | - Sang Hoon Park
- Department of Internal Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Freddy E Escorcia
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National institutes of Health, Bethesda, MD, USA
| | - Joon-Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National institutes of Health, Bethesda, MD, USA
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Technology, Institute for Bio-medical Convergence Science and Technology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Tattoli I, Mathew AR, Verrienti A, Pallotta L, Severi C, Andreola F, Cavallucci V, Giorgi M, Massimi M, Bencini L, Fidaleo M. The Interplay between Liver and Adipose Tissue in the Onset of Liver Diseases: Exploring the Role of Vitamin Deficiency. Cells 2024; 13:1631. [PMID: 39404394 PMCID: PMC11475612 DOI: 10.3390/cells13191631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
The deficiency of vitamins, a condition known as "hidden hunger", causes comprehensive pathological states. Research over the years has identified a relationship between liver diseases and hypovitaminosis or defects in vitamin metabolism. The exact mechanisms remain elusive; however, the crucial involvement of specific vitamins in metabolic functions, alongside the reclassification of liver disease as metabolic dysfunction-associated steatotic liver disease (MASLD), has prompted researchers to investigate the potential cause-effect dynamics between vitamin deficiency and liver disease. Moreover, scientists are increasingly investigating how the deficiency of vitamins might disrupt specific organ crosstalk, potentially contributing to liver disease. Although the concept of a dysmetabolic circuit linking adipose tissue and the liver, leading to liver disease, has been discussed, the possible involvement of vitamin deficiency in this axis is a relatively recent area of study, with numerous critical aspects yet to be fully understood. In this review, we examine research from 2019 to July 2024 focusing on the possible link between liver-adipose tissue crosstalk and vitamin deficiency involved in the onset and progression of non-alcoholic fatty liver disease (NAFLD). Studies report that vitamin deficiency can affect the liver-adipose tissue axis, mainly affecting the regulation of systemic energy balance and inflammation.
Collapse
Affiliation(s)
- Ivan Tattoli
- Oncology General Surgery, Azienda Ospedaliero Universitaria Careggi, 50139 Florence, Italy; (I.T.); (L.B.)
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (M.G.)
| | - Aimee Rachel Mathew
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (M.G.)
| | - Antonella Verrienti
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00161 Rome, Italy; (A.V.); (L.P.); (C.S.)
| | - Lucia Pallotta
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00161 Rome, Italy; (A.V.); (L.P.); (C.S.)
| | - Carola Severi
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00161 Rome, Italy; (A.V.); (L.P.); (C.S.)
| | - Fausto Andreola
- Liver Failure Group, Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK;
| | - Virve Cavallucci
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy;
| | - Mauro Giorgi
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (M.G.)
| | - Mara Massimi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Lapo Bencini
- Oncology General Surgery, Azienda Ospedaliero Universitaria Careggi, 50139 Florence, Italy; (I.T.); (L.B.)
| | - Marco Fidaleo
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (M.G.)
- Research Center for Nanotechnology for Engineering of Sapienza (CNIS), Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
13
|
Tripathi M, Gauthier K, Sandireddy R, Zhou J, Guptta P, Sakthivel S, Teo WW, Naing YT, Arul K, Tikno K, Park SH, Wu Y, Wang L, Bay BH, Sun L, Giguere V, Chow PKH, Ghosh S, McDonnell DP, Yen PM, Singh BK. Esrra regulates Rplp1-mediated translation of lysosome proteins suppressed in metabolic dysfunction-associated steatohepatitis and reversed by alternate day fasting. Mol Metab 2024; 87:101997. [PMID: 39032642 PMCID: PMC11327444 DOI: 10.1016/j.molmet.2024.101997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/03/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024] Open
Abstract
OBJECTIVE Currently, little is known about the mechanism(s) regulating global and specific protein translation during metabolic dysfunction-associated steatohepatitis (MASH; previously known as non-alcoholic steatohepatitis, NASH). METHODS Unbiased label-free quantitative proteome, puromycin-labelling and polysome profiling were used to understand protein translation activity in vitro and in vivo. RESULTS We observed a global decrease in protein translation during lipotoxicity in human primary hepatocytes, mouse hepatic AML12 cells, and livers from a dietary mouse model of MASH. Interestingly, proteomic analysis showed that Rplp1, which regulates ribosome and translation pathways, was one of the most downregulated proteins. Moreover, decreased Esrra expression and binding to the Rplp1 promoter, diminished Rplp1 gene expression during lipotoxicity. This, in turn, reduced global protein translation and Esrra/Rplp1-dependent translation of lysosome (Lamp2, Ctsd) and autophagy (sqstm1, Map1lc3b) proteins. Of note, Esrra did not increase its binding to these gene promoters or their gene transcription, confirming its regulation of their translation during lipotoxicity. Notably, hepatic Esrra-Rplp1-dependent translation of lysosomal and autophagy proteins also was impaired in MASH patients and liver-specific Esrra knockout mice. Remarkably, alternate day fasting induced Esrra-Rplp1-dependent expression of lysosomal proteins, restored autophagy, and reduced lipotoxicity, inflammation, and fibrosis in hepatic cell culture and in vivo models of MASH. CONCLUSIONS Esrra regulation of Rplp1-mediated translation of lysosome/autolysosome proteins was downregulated during MASH. Alternate day fasting activated this novel pathway and improved MASH, suggesting that Esrra and Rplp1 may serve as therapeutic targets for MASH. Our findings also provided the first example of a nuclear hormone receptor, Esrra, to not only regulate transcription but also protein translation, via induction of Rplp1.
Collapse
Affiliation(s)
- Madhulika Tripathi
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Karine Gauthier
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie 69364 Lyon Cedex 07, France
| | - Reddemma Sandireddy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Jin Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Priyanka Guptta
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Suganya Sakthivel
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Wei Wen Teo
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Yadanar Than Naing
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Kabilesh Arul
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Keziah Tikno
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Sung-Hee Park
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C238A Levine Science Research Center, Durham, NC 27710, USA
| | - Yajun Wu
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS 117594, Singapore
| | - Lijin Wang
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore; Pennington Biomedical Research Center, Laboratory of Bioinformatics and Computational Biology, Baton Rouge, LA 70808, USA
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS 117594, Singapore
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Vincent Giguere
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, Québec H3A 1A3, Canada
| | - Pierce K H Chow
- Dept of Surgery, Singapore General Hospital and Dept. of Surgical Oncology, National Cancer Centre 169608, Singapore
| | - Sujoy Ghosh
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore; Pennington Biomedical Research Center, Laboratory of Bioinformatics and Computational Biology, Baton Rouge, LA 70808, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C238A Levine Science Research Center, Durham, NC 27710, USA
| | - Paul M Yen
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore; Duke Molecular Physiology Institute and Dept. of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Brijesh K Singh
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore.
| |
Collapse
|
14
|
Raza S, Rajak S, Yen PM, Sinha RA. Autophagy and hepatic lipid metabolism: mechanistic insight and therapeutic potential for MASLD. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:19. [PMID: 39100919 PMCID: PMC11296953 DOI: 10.1038/s44324-024-00022-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) originates from a homeostatic imbalance in hepatic lipid metabolism. Increased fat deposition in the liver of people suffering from MASLD predisposes them to develop further metabolic derangements, including diabetes mellitus, metabolic dysfunction-associated steatohepatitis (MASH), and other end-stage liver diseases. Unfortunately, only limited pharmacological therapies exist for MASLD to date. Autophagy, a cellular catabolic process, has emerged as a primary mechanism of lipid metabolism in mammalian hepatocytes. Furthermore, preclinical studies with autophagy modulators have shown promising results in resolving MASLD and mitigating its progress into deleterious liver pathologies. In this review, we discuss our current understanding of autophagy-mediated hepatic lipid metabolism, its therapeutic modulation for MASLD treatment, and current limitations and scope for clinical translation.
Collapse
Affiliation(s)
- Sana Raza
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014 India
| | - Sangam Rajak
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014 India
| | - Paul M. Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, 169857 Singapore
| | - Rohit A. Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014 India
| |
Collapse
|
15
|
Li M, Zhang J, Li Z, Xu Z, Qian S, Tay LJ, Zhang Z, Yang F, Huang Y. The role and mechanism of SUMO modification in liver disease. Biomed Pharmacother 2024; 177:116898. [PMID: 38878635 DOI: 10.1016/j.biopha.2024.116898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 07/28/2024] Open
Abstract
Liver disease affects millions of people in the world, and China has the highest prevalence of liver disease in the world. Small ubiquitin-related modifier (SUMO) modification is a highly conserved post-translational modification of proteins. They are widely expressed in a variety of tissues, including the heart, liver, kidney and lung. SUMOylation of protein plays a key role in the occurrence and development of liver disease. Therefore, this study reviewed the effects of SUMO protein on non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), viral hepatitis, hepatic fibrosis (HF), hepatocellular carcinoma (HCC), and other liver diseases to provide novel strategies for targeted treatment of liver disease.
Collapse
Affiliation(s)
- Mengxue Li
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Jingrong Zhang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Zihao Li
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Zhou Xu
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Shishun Qian
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Lynn Jia Tay
- School of International Education, Anhui Medical University, Hefei 230032, China
| | - Ziwen Zhang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Furong Yang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China.
| | - Yan Huang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China; School of International Education, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
16
|
Sandireddy R, Sakthivel S, Gupta P, Behari J, Tripathi M, Singh BK. Systemic impacts of metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH) on heart, muscle, and kidney related diseases. Front Cell Dev Biol 2024; 12:1433857. [PMID: 39086662 PMCID: PMC11289778 DOI: 10.3389/fcell.2024.1433857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is the most common liver disorder worldwide, with an estimated global prevalence of more than 31%. Metabolic dysfunction-associated steatohepatitis (MASH), formerly known as non-alcoholic steatohepatitis (NASH), is a progressive form of MASLD characterized by hepatic steatosis, inflammation, and fibrosis. This review aims to provide a comprehensive analysis of the extrahepatic manifestations of MASH, focusing on chronic diseases related to the cardiovascular, muscular, and renal systems. A systematic review of published studies and literature was conducted to summarize the findings related to the systemic impacts of MASLD and MASH. The review focused on the association of MASLD and MASH with metabolic comorbidities, cardiovascular mortality, sarcopenia, and chronic kidney disease. Mechanistic insights into the concept of lipotoxic inflammatory "spill over" from the MASH-affected liver were also explored. MASLD and MASH are highly associated (50%-80%) with other metabolic comorbidities such as impaired insulin response, type 2 diabetes, dyslipidemia, hypertriglyceridemia, and hypertension. Furthermore, more than 90% of obese patients with type 2 diabetes have MASH. Data suggest that in middle-aged individuals (especially those aged 45-54), MASLD is an independent risk factor for cardiovascular mortality, sarcopenia, and chronic kidney disease. The concept of lipotoxic inflammatory "spill over" from the MASH-affected liver plays a crucial role in mediating the systemic pathological effects observed. Understanding the multifaceted impact of MASH on the heart, muscle, and kidney is crucial for early detection and risk stratification. This knowledge is also timely for implementing comprehensive disease management strategies addressing multi-organ involvement in MASH pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | - Madhulika Tripathi
- Cardiovascular and Metabolic Disorders Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Brijesh Kumar Singh
- Cardiovascular and Metabolic Disorders Research Program, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
17
|
Yu W, Chen J, Jin S, Fan X, Cai X. Identification and Validation of Glycosylation-Related Genes in Obesity and MASH: Insights from Human Liver Samples and a High-Fat Diet Mouse Model. Pharmgenomics Pers Med 2024; 17:363-381. [PMID: 38983907 PMCID: PMC11232960 DOI: 10.2147/pgpm.s463608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/12/2024] [Indexed: 07/11/2024] Open
Abstract
Background Obesity is reaching epidemic proportions in the developed world. The biosynthesis and degradation of human glycoproteins take place at the highest level in the liver. However, the association between glycosylation and the factors affecting obesity and metabolism-associated steatohepatitis (MASH) is still unclear. Materials and Methods Gene expression data of liver samples from obese patients were retrieved from GSE83452 and GSE89632 databases. Difference analysis and machine learning were used to identify hub genes involved in glycosylation and associated with the response of weight loss treatment. A total of 7 glycosylation-related hub genes were identified and then subjected to correlation analysis, immune cells infiltration analysis and ROC (Receiver Operating Characteristic) analysis. We also evaluated the potential function of 7 hub genes in obesity patients. MASH mice were used to validate the glycosylation-related hub genes. Results A total of 25 overlapped glycosylation-related genes were identified by DEGs analysis. ACER2, STX17, ARF5, GPC4, ENTPD5, NANP, and DPY19L2 were identified as hub genes. Among these hub genes, ACER2, STX17, ARF5, and ENTPD5 were also differential expressed in MASH patients. ENTPD5 showed increased transcription in obese MASH mice. Conclusion The current study identified seven glycosylation-related genes, ACER2, STX17, ARF5, GPC4, ENTPD5, NANP, and DPY19L2, that might play key roles in the development of obesity. ENTPD5 might play a key role in the development of MASH. These findings provide fresh perspectives for expanding the investigation of obesity and MASH.
Collapse
Affiliation(s)
- Weihua Yu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jionghuang Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Shengxi Jin
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiaoxiao Fan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiujun Cai
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
18
|
Yang M, Wang D, Wang X, Mei J, Gong Q. Role of Folate in Liver Diseases. Nutrients 2024; 16:1872. [PMID: 38931227 PMCID: PMC11206401 DOI: 10.3390/nu16121872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Folate is a water-soluble B vitamin involved in the synthesis of purines and pyrimidines and is one of the essential vitamins for human growth and reproduction. Folate deficiency due to low dietary intake, poor absorption of folate, and alterations in folate metabolism due to genetic defects or drug interactions significantly increases the risk of diseases such as neural tube defects, cardiovascular disease, cancer, and cognitive dysfunction. Recent studies have shown that folate deficiency can cause hyperhomocysteinemia, which increases the risk of hypertension and cardiovascular disease, and that high homocysteine levels are an independent risk factor for liver fibrosis and cirrhosis. In addition, folate deficiency results in increased secretion of pro-inflammatory factors and impaired lipid metabolism in the liver, leading to lipid accumulation in hepatocytes and fibrosis. There is substantial evidence that folate deficiency contributes to the development and progression of a variety of liver diseases, including non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), alcoholic liver disease (ALD), viral hepatitis, hepatic fibrosis, and liver cancer. Here we review key studies on the role of folate in the pathophysiology of liver diseases, summarize the current status of studies on folate in the treatment of liver diseases, and speculate that folate may be a potential therapeutic target for liver diseases.
Collapse
Affiliation(s)
- Minlan Yang
- School of Medicine, Yangtze University, Jingzhou 434020, China
| | | | | | | | - Quan Gong
- School of Medicine, Yangtze University, Jingzhou 434020, China
| |
Collapse
|
19
|
India-Aldana S, Midya V, Betanzos-Robledo L, Yao M, Alcalá C, Andra SS, Arora M, Calafat AM, Chu J, Deierlein A, Estrada-Gutierrez G, Jagani R, Just AC, Kloog I, Landero J, Oulhote Y, Walker RW, Yelamanchili S, Baccarelli AA, Wright RO, Téllez Rojo MM, Colicino E, Cantoral A, Valvi D. Metabolism-Disrupting Chemical Mixtures during Pregnancy, Folic Acid Supplementation, and Liver Injury in Mother-Child Pairs. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.13.24308903. [PMID: 38947077 PMCID: PMC11213105 DOI: 10.1101/2024.06.13.24308903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background and Aims Scarce knowledge about the impact of metabolism-disrupting chemicals (MDCs) on liver injury limits opportunities for intervention. We evaluated pregnancy MDC-mixture associations with liver injury and effect modification by folic acid (FA) supplementation in mother-child pairs. Methods We studied ∼200 mother-child pairs from the Mexican PROGRESS cohort, with measured 43 MDCs during pregnancy (estimated air pollutants, blood/urine metals or metalloids, urine high- and low-molecular-weight phthalate [HMWPs, LMWPs] and organophosphate-pesticide [OP] metabolites), and serum liver enzymes (ALT, AST) at ∼9 years post-parturition. We defined liver injury as elevated liver enzymes in children, and using established clinical scores for steatosis and fibrosis in mothers (i.e., AST:ALT, FLI, HSI, FIB-4). Bayesian Weighted Quantile Sum regression assessed MDC-mixture associations with liver injury outcomes. We further examined chemical-chemical interactions and effect modification by self-reported FA supplementation. Results In children, many MDC-mixtures were associated with liver injury outcomes. Per quartile HMWP-mixture increase, ALT increased by 10.1% (95%CI: 1.67%, 19.4%) and AST by 5.27% (95% CI: 0.80%, 10.1%). LMWP-mixtures and air pollutant-mixtures were associated with higher AST and ALT, respectively. Air pollutant and non-essential metal/element associations with liver enzymes were attenuated by maternal cobalt blood concentrations ( p -interactions<0.05). In mothers, only the LMWP-mixture was associated with liver injury [OR=1.53 (95%CI: 1.01, 2.28) for HSI>36, and OR=1.62 (95%CI: 1.05, 2.49) for AST:ALT<1]. In mothers and children, most associations were attenuated (null) at FA supplementation≥600mcg/day ( p -interactions<0.05). Conclusions Pregnancy MDC exposures may increase liver injury risk, particularly in children. These associations may be attenuated by higher FA supplementation and maternal cobalt levels.
Collapse
|
20
|
Ji Q, Zhang F, Su Q, He T, Wu Z, Zhu K, Chen X, Wang Z, Hou S, Gui L. Effect of supplementing lysins and methionine to low-protein diets on growth performance, hepatic antioxidant capacity, immune status, and glycolytic activity of tibetan sheep. BMC Genomics 2024; 25:557. [PMID: 38834972 DOI: 10.1186/s12864-024-10480-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 05/30/2024] [Indexed: 06/06/2024] Open
Abstract
Reducing the levels of dietary protein is an effective nutritional approach in lowering feed cost and nitrogen emissions in ruminants. The purpose of this study was to evaluate the effects of dietary Lys/Met ratio in a low protein diet (10%, dry matter basis) on the growth performance and hepatic function (antioxidant capacity, immune status, and glycolytic activity) in Tibetan lambs. Ninety two-month-old rams with an average weight of 15.37 ± 0.92 kg were randomly assigned to LP-L (dietary Lys/Met = 1:1), LP-M (dietary Lys/Met = 2:1) and LP-H (dietary Lys/Met = 3:1) treatments. The trial was conducted over 100 d, including 10 d of adaption to the diets. Hepatic phenotypes, antioxidant capacity, immune status, glycolytic activity and gene expression profiling was detected after the conclusion of the feeding trials. The results showed that the body weight was higher in the LP-L group when compared to those on the LP-M group (P < 0.05). In addition, the activities of the catalase (CAT) and glutathione peroxidase (GSH-Px) in the LP-L group were significantly increased compared with the LP-M group (P < 0.05), while the malondialdehyde (MDA) levels in LP-H group were significantly decreased (P < 0.05). Compared with LP-H group, both hepatic glycogen (P < 0.01) and lactate dehydrogenase (LDH) (P < 0.05) were significantly elevated in LP-L group. For the LP-L group, the hepatocytes were arranged radially with the central vein in the center, and hepatic plates exhibited tight arrangement. Transcriptome analysis identified 29, 179, and 129 differentially expressed genes (DEGs) between the LP-M vs. LP-L, LP-H vs. LP-M, and LP-H vs. LP-L groups, respectively (Q-values < 0.05 and |log2Fold Change| > 1). Gene Ontology (GO) and correlation analyses showed that in the LP-L group, core genes (C1QA and JUNB) enriched in oxidoreductase activity were positively correlated with antioxidant indicators, while the MYO9A core gene enriched in the immune response was positively associated with immune indicators, and core genes enriched in molecular function (PDK3 and PDP2) were positively correlated with glycolysis indicators. In summary, low-protein diet with a low Lys/Met ratio (1:1) could reduce the hepatic oxidative stress and improve the glycolytic activity by regulating the expression of related genes of Tibetan sheep.
Collapse
Affiliation(s)
- Qiurong Ji
- Qinghai University College of Agriculture and Animal Husbandry, Xining, 810016, China
| | - Fengshuo Zhang
- Qinghai University College of Agriculture and Animal Husbandry, Xining, 810016, China
| | - Quyangangmao Su
- Qinghai University College of Agriculture and Animal Husbandry, Xining, 810016, China
| | - Tingli He
- Qinghai University College of Agriculture and Animal Husbandry, Xining, 810016, China
| | - Zhenling Wu
- Qinghai University College of Agriculture and Animal Husbandry, Xining, 810016, China
| | - Kaina Zhu
- Qinghai University College of Agriculture and Animal Husbandry, Xining, 810016, China
| | - Xuan Chen
- Qinghai University College of Agriculture and Animal Husbandry, Xining, 810016, China
| | - Zhiyou Wang
- Qinghai University College of Agriculture and Animal Husbandry, Xining, 810016, China
| | - Shengzhen Hou
- Qinghai University College of Agriculture and Animal Husbandry, Xining, 810016, China
| | - Linsheng Gui
- Qinghai University College of Agriculture and Animal Husbandry, Xining, 810016, China.
| |
Collapse
|
21
|
Suzuki A, Henao R, Reed MC, Nijhout HF, Tripathi M, Singh BK, Yen PM, Diehl AM, Abdelmalek MF. Lower hepatic CBS and PEMT expression in advanced NAFLD: inferencing strategies to lower homocysteine with a mathematical model. METABOLISM AND TARGET ORGAN DAMAGE 2024; 4. [DOI: 10.20517/mtod.2024.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Aim: Hepatic homocysteine (Hcy) accumulation promotes inflammation and fibrosis in experimental nonalcoholic fatty liver disease (NAFLD), while vitamin B12 and folate reduce hepatic Hcy and protect animals from nonalcoholic steatohepatitis. This suggests clinical implications for preventing/treating patients with NAFLD. Given the known sex-specific regulation of one-carbon metabolism (OCM), the response to various OCM cofactors may vary by sex and reproductive status. We aimed to strategize an effective Hcy-lowering treatment in broader NAFLD patients while discerning disparities in treatment responses.
Methods: We analyzed existing hepatic microarray data relevant to Hcy metabolism with clinical and histologic data from patients with NAFLD (N = 82), while considering potential age/sex disparities. Additionally, we performed computer simulation analyses using a mathematical model of OCM to predict hepatic Hcy-lowering effects of OCM cofactors by sex.
Results: Of 82 patients with NAFLD, 98% had at least one metabolic feature [i.e., metabolic dysfunction-associated steatotic liver disease (MASLD)]. Lower hepatic gene expressions of cystathionine-beta synthase (CBS ) and phosphatidyl- ethanolamine N-methyltransferase (PEMT ) were associated with more severe fibrosis in NAFLD, while sub-analysis suggested possible variations by age and sex. The simulation analysis demonstrated sex differences in the Hcy-lowering effects of the OCM cofactors (vitamins B6 and B12, folate, and betaine), with the combination of these cofactors consistently showing the maximum Hcy-lowering effect in both sexes.
Conclusion: We theorize that the combination of OCM cofactors would maximize Hcy-lowering effects in the broader MASLD population. Our findings also underscore the importance of considering sex and age in designing future studies on homocysteine metabolism.
Collapse
|
22
|
Guo T, Zhou L, Xiong M, Xiong J, Huang J, Li Y, Zhang G, Chen G, Wang Z, Xiao T, Hu D, Bao A, Zhang Z. N-homocysteinylation of DJ-1 promotes neurodegeneration in Parkinson's disease. Aging Cell 2024; 23:e14124. [PMID: 38380563 PMCID: PMC11113254 DOI: 10.1111/acel.14124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/31/2023] [Accepted: 02/11/2024] [Indexed: 02/22/2024] Open
Abstract
DJ-1, also known as Parkinson's disease protein 7 (Park7), is a multifunctional protein that regulates oxidative stress and mitochondrial function. Dysfunction of DJ-1 is implicated in the pathogenesis of Parkinson's disease (PD). Hyperhomocysteinemia is associated with an increased risk of PD. Here we show that homocysteine thiolactone (HTL), a reactive thioester of homocysteine (Hcy), covalently modifies DJ-1 on the lysine 182 (K182) residue in an age-dependent manner. The N-homocysteinylation (N-hcy) of DJ-1 abolishes its neuroprotective effect against oxidative stress and mitochondrial dysfunction, exacerbating cell toxicity. Blocking the N-hcy of DJ-1 restores its protective effect. These results indicate that the N-hcy of DJ-1 abolishes its neuroprotective effect and promotes the progression of PD. Inhibiting the N-hcy of DJ-1 may exert neuroprotective effect against PD.
Collapse
Affiliation(s)
- Tao Guo
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Lingyan Zhou
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Min Xiong
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jing Xiong
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Juan Huang
- Department of NeurologySecond Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Yiming Li
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Guoxin Zhang
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Guiqin Chen
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhi‐Hao Wang
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Tingting Xiao
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Dan Hu
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Anyu Bao
- Department of Clinical LaboratoryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhentao Zhang
- Department of NeurologyRenmin Hospital of Wuhan UniversityWuhanChina
- TaiKang Center for Life and Medical SciencesWuhan UniversityWuhanChina
| |
Collapse
|
23
|
Fu Y, Wang Z, Qin H. Examining the Pathogenesis of MAFLD and the Medicinal Properties of Natural Products from a Metabolic Perspective. Metabolites 2024; 14:218. [PMID: 38668346 PMCID: PMC11052500 DOI: 10.3390/metabo14040218] [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: 03/17/2024] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD), characterized primarily by hepatic steatosis, has become the most prevalent liver disease worldwide, affecting approximately two-fifths of the global population. The pathogenesis of MAFLD is extremely complex, and to date, there are no approved therapeutic drugs for clinical use. Considerable evidence indicates that various metabolic disorders play a pivotal role in the progression of MAFLD, including lipids, carbohydrates, amino acids, and micronutrients. In recent years, the medicinal properties of natural products have attracted widespread attention, and numerous studies have reported their efficacy in ameliorating metabolic disorders and subsequently alleviating MAFLD. This review aims to summarize the metabolic-associated pathological mechanisms of MAFLD, as well as the natural products that regulate metabolic pathways to alleviate MAFLD.
Collapse
Affiliation(s)
| | | | - Hong Qin
- Department of Nutrition and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha 410006, China; (Y.F.); (Z.W.)
| |
Collapse
|
24
|
Huang J, Li J, Peng Y, Cui T, Guo J, Duan S, Zhou K, Huang S, Chen J, Yi Q, Qiu M, Chen T, Wu X, Ma C, Zhang Z, Zheng Y, Tang X, Pang Y, Zhang L, Zhong C, Gao Y. The lack of PPARα exacerbated the progression of non-alcoholic steatohepatitis in mice with spleen deficiency syndrome by triggering an inflammatory response. Front Immunol 2024; 15:1381340. [PMID: 38633246 PMCID: PMC11021588 DOI: 10.3389/fimmu.2024.1381340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 03/14/2024] [Indexed: 04/19/2024] Open
Abstract
Background In addition to abnormal liver inflammation, the main symptoms of non-alcoholic steatohepatitis (NASH) are often accompanied by gastrointestinal digestive dysfunction, consistent with the concept of spleen deficiency (SD) in traditional Chinese medicine. As an important metabolic sensor, whether peroxisome proliferator-activated receptor alpha (PPARα) participates in regulating the occurrence and development of NASH with SD (NASH-SD) remains to be explored. Methods Clinical liver samples were collected for RNA-seq analysis. C57BL/6J mice induced by folium sennae (SE) were used as an SD model. qPCR analysis was conducted to evaluate the inflammation and metabolic levels of mice. PPARα knockout mice (PPARαko) were subjected to SE and methionine-choline-deficient (MCD) diet to establish the NASH-SD model. The phenotype of NASH and the inflammatory indicators were measured using histopathologic analysis and qPCR as well. Results The abnormal expression of PPARα signaling, coupled with metabolism and inflammation, was found in the results of RNA-seq analysis from clinical samples. SD mice showed a more severe inflammatory response in the liver evidenced by the increases in macrophage biomarkers, inflammatory factors, and fibrotic indicators in the liver. qPCR results also showed differences in PPARα between SD mice and control mice. In PPARαko mice, further evidence was found that the lack of PPARα exacerbated the inflammatory response phenotype as well as the lipid metabolism disorder in NASH-SD mice. Conclusion The abnormal NR signaling accelerated the vicious cycle between lipotoxicity and inflammatory response in NAFLD with SD. Our results provide new evidence for nuclear receptors as potential therapeutic targets for NAFLD with spleen deficiency.
Collapse
Affiliation(s)
- Jiawen Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiayu Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuan Peng
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Tianqi Cui
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingyi Guo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Siwei Duan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kaili Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shangyi Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiabing Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qincheng Yi
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Qiu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tingting Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoqin Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chenlu Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziyi Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Zheng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xi Tang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanqing Pang
- Department of Phase I Clinical Research Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Lei Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chong Zhong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Gao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
25
|
Shi X, Xiong Y, Song H, Rong F, Tang N, Zhu L, Li S, Wang J, Zhang L, You S, Ji G, Liu B, Wu N. Progress and hotspot of diet or exercise therapy in the treatment of non-alcoholic fatty liver disease. Front Nutr 2024; 11:1326092. [PMID: 38628270 PMCID: PMC11018916 DOI: 10.3389/fnut.2024.1326092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/15/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction The primary treatment for non-alcoholic fatty liver disease (NAFLD) is modifying lifestyle through dietary or exercise interventions. In recent decades, it has received increasing attention. However, the lack of bibliometric analysis has posed a challenge for researchers seeking to understand the overall trends in this field. Methods As of February 3rd, 2024, 876 articles on treating NAFLD through diet or exercise therapy from 2013 to 2023 had been retrieved. Two software tools, VOSviewer and CiteSpace, were utilized to analyze the growth of publications, countries, institutions, authors, journals, citations, and keywords. Additionally, the keywords with strong citation burstiness were identified to determine the changes and future trends of research hotspots in this field. Results China had the highest number of articles, followed by the United States and South Korea. Yonsei University and Nutrients were the institutions and journals with the most significant contributions. Professor Younossi Zobair M, from the United States, is the most prolific author in this field. Through analyzing the keywords, three research hotspots were identified: research on the pathogenesis of NAFLD, research on the treatment modalities of NAFLD, and research on the risk factors and diagnosis methods of NAFLD. In recent years, the research emphasis in this field has changed, suggesting that future research will focus on two frontier keywords: "oxidative stress" and "aerobic capacity." Conclusion In the past eleven years, the attention in this field was still rising, and the authors, journals, countries and so on had formed a considerable cooperative relationship. There were also many highly influential and productive researchers in this field. It is speculated that new research will continue around "aerobic exercise" and "oxidative stress" in the future.
Collapse
Affiliation(s)
- Xinyu Shi
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yalan Xiong
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hualing Song
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fen Rong
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Nan Tang
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Leping Zhu
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shenyu Li
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianying Wang
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Zhang
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shengfu You
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Baocheng Liu
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Na Wu
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
26
|
Li J, Tian S, Ci B, Xi Y, Deng X. Serum vitamins and homocysteine levels in autoimmune liver disease: A systematic review and meta-analysis. Immun Inflamm Dis 2024; 12:e1258. [PMID: 38652023 PMCID: PMC11037259 DOI: 10.1002/iid3.1258] [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: 01/17/2024] [Revised: 03/24/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
OBJECTIVE Vitamins and homocysteine (Hcy) are involved in liver metabolism and related to the pathogenesis of autoimmune liver disease (AILD), but consensus is lacking. This study aims to systematically summarize relevant evidence to clarify the association of serum vitamins and Hcy levels with AILD. METHODS The English and Chinese literature was searched until August 29, 2023. Studies were included if they were observational studies of investigating serum vitamins and Hcy levels in patients with AILD and their healthy comparisons. Quality assessment was performed by using the Newcastle-Ottawa Scale, and a meta-analysis was conducted using ReviewManager 5.3. The protocol was registered in the international prospective register of systematic reviews (PROSPERO), with registration number CRD42023455367. RESULTS A total of 25 case-control studies comprising 3487 patients (1673 patients and 1814 healthy controls) were included for analysis. There were 548 autoimmune hepatitis (AIH) cases, 1106 primary biliary cholangitis (PBC) cases, and 19 primary sclerosing cholangitis (PSC) cases. We found that serum A and E were decreased in both AIH and PBC/PSC; but vitamin C was reduced only in patients with PBC, not AIH. In addition, decreased content of 25(OH)D3 was found in both AIH and PBC. However, levels of 25(OH)D did not differ between the patients and controls, and were independent of disease types and the country. Only one study that met the inclusion criteria reported vitamin B6, B9, B12, and Hcy changes, and found that vitamin B6 and B9 were significantly decreased in patients with PBC, while serum vitamin B12 and Hcy levels were significantly elevated in them. One eligible study each confirmed a reduction in plasma vitamin K1 and 1,25(OH)2D3 in patients with PBC. CONCLUSION Most vitamins are deficient in AILD, so appropriate vitamin supplementation should be necessary. Further studies with larger sample sizes are needed to validate these findings.
Collapse
Affiliation(s)
- Jiahuan Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Shan Tian
- Department of Infectious Diseases, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Bai Ci
- Division of Gastroenterology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuwen Xi
- Division of Gastroenterology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoling Deng
- Division of Gastroenterology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
27
|
Qiu Y, Li C, Huang Y, Wu C, Li F, Zhang X, Xia D. Exploring the causal associations of micronutrients on urate levels and the risk of gout: A Mendelian randomization study. Clin Nutr 2024; 43:1001-1012. [PMID: 38484526 DOI: 10.1016/j.clnu.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND & AIMS Growing evidence has indicated a potential association between micronutrient levels, urate levels, and the risk of gout. However, the causal association underlying these associations still remains uncertain. Previous observational studies and randomized controlled trials investigating the association between micronutrients, urate levels, and the risk of gout have been limited in their scope and depth. The aim of this study was to utilize Mendelian randomization (MR) to investigate the causal associations between genetically predicted micronutrient levels, urate levels, and the risk of gout. METHODS In this study, we conducted a comprehensive examination of 10 specific micronutrients (vitamin B6, vitamin B12, vitamin C, vitamin D, folate, calcium, iron, copper, zinc, and selenium) as potential exposures. Two-sample MR analyses were performed to explore their causal associations with urate levels and the risk of gout. In these analyses, gout data were collected from the Global Biobank Meta-Analysis Initiative (N = 1,069,839, N cases = 30,549) and urate levels data from CKDGen Consortium (N = 288,649) by utilizing publicly available summary statistics from independent cohorts of European ancestry. We performed inverse-variance weighted MR analyses as main analyses, along with a range of sensitivity analyses, such as MR-Egger, weighted median, simple mode, weighted mode, Steiger filtering, MR-PRESSO, and Radial MR analysis, to ensure the robustness of our findings. RESULTS The results of our study indicate that there were negative associations between serum vitamin B12 and urate levels, as well as serum folate and the risk of gout. Specifically, we found a negative association between vitamin B12 levels and urate levels, with a β coefficient of -0.324 (95% CI -0.0581 to -0.0066, P = 0.0137) per one standard deviation (SD) increase. Similarly, a negative association was observed between folate levels and gout risk, with an odds ratio of 0.8044 (95% CI 0.6637 to 0.9750, P = 0.0265) per one SD increase. On the other hand, we identified positive associations between serum calcium levels and both urate levels and the risk of gout. Specifically, there was a positive association between serum calcium levels and urate levels (β coefficient: 0.0994, 95% CI 0.0519 to 0.1468, P = 4.11E-05) per one SD increase. Furthermore, a positive association was found between serum calcium levels and the risk of gout, with an odds ratio of 1.1479 (95% CI 1.0460 to 1.2598, P = 0.0036) per one SD increase. These findings were robust in extensive sensitivity analyses. By employing MR-PRESSO and Radial MR to eliminate outliers, the observed associations have been reinforced. No clear associations were found between the other micronutrients and the urate levels, as well as the risk of gout. CONCLUSION Our findings provided evidence that there were negative associations between serum vitamin B12 and urate levels, as well as serum folate and the risk of gout, while positive associations existed between the serum calcium levels and urate levels, as well as the risk of gout.
Collapse
Affiliation(s)
- Yu Qiu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Cantao Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenxi Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fenfen Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaoxi Zhang
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Daozong Xia
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
28
|
Deng Y, Hu M, Huang S, Fu N. Molecular mechanism and therapeutic significance of essential amino acids in metabolically associated fatty liver disease. J Nutr Biochem 2024; 126:109581. [PMID: 38219809 DOI: 10.1016/j.jnutbio.2024.109581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/01/2024] [Accepted: 01/06/2024] [Indexed: 01/16/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD), also known as metabolically associated fatty liver disease (MAFLD), is a systemic metabolic disease characterized by lipid accumulation in the liver, lipid toxicity, insulin resistance, intestinal dysbiosis, and inflammation that can progress from simple steatosis to nonalcoholic steatohepatitis (NASH) and even cirrhosis or cancer. It is the most prevalent illness threatening world health. Currently, there are almost no approved drug interventions for MAFLD, mainly dietary changes and exercise to control weight and regulate metabolic disorders. Meanwhile, the metabolic pathway involved in amino acid metabolism also influences the onset and development of MAFLD in the body, and most amino acid metabolism takes place in the liver. Essential amino acids are those amino acids that must be supplemented from outside the diet and that cannot be synthesized in the body or cannot be synthesized at a rate sufficient to meet the body's needs, including leucine, isoleucine, valine (collectively known as branched-chain amino acids), tryptophan, phenylalanine (which are aromatic amino acids), histidine, methionine, threonine and lysine. The metabolic balance of the body is closely linked to these essential amino acids, and essential amino acids are closely linked to the pathophysiological process of MAFLD. In this paper, we will focus on the metabolism of essential amino acids in the body and further explore the therapeutic strategies for MAFLD based on the studies conducted in recent years.
Collapse
Affiliation(s)
- Yuting Deng
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Mengsi Hu
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Shufang Huang
- The Affiliated Nanhua Hospital, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China.
| | - Nian Fu
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China; The Affiliated Nanhua Hospital, Institute of Clinical Research, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China.
| |
Collapse
|
29
|
Jia Y, Li J, Wang Y, Ma Y, Chen L, Zhang H, Xue M, Liang H. Folic Acid Rescues Dopaminergic Neurons in MPTP-Induced Mice by Inhibiting the NLRP3 Inflammasome and Ameliorating Mitochondrial Impairment. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:5734-5745. [PMID: 38453725 DOI: 10.1021/acs.jafc.3c06337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Parkinson's disease (PD) is marked by the degeneration of dopaminergic neurons of the substantia nigra (SN), with neuroinflammation and mitochondrial dysfunction being key contributors. The neuroprotective potential of folic acid (FA) in the dopaminergic system of PD was assessed in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model. MPTP (20 mg/kg of body weight) was administered to C57BL/6J mice to simulate PD symptoms followed by FA treatment (5 mg/kg of body weight). Behavioral tests, pole, rotarod, and open-field tests, evaluated motor function, while immunohistochemistry, ELISA, RT-qPCR, and Western blotting quantified neuroinflammation, oxidative stress markers, and mitochondrial function. FA supplementation considerably improved motor performance, reduced homocysteine levels and mitigated oxidative damage in the SN. The FA-attenuated activation of the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome lessened glial cell activity and reduced neuroinflammation. At the molecular level, FA reduced DNA damage, downregulated phosphorylated p53, and induced the expression of peroxisome proliferator-activated receptor α coactivator 1α (PGC-1α), enhancing mitochondrial function. Therefore, FA exerts neuroprotection in MPTP-induced PD by inhibiting neuroinflammation via NLRP3 inflammasome suppression and promoting mitochondrial integrity through the p53-PGC-1α pathway. Notable limitations of our study include its reliance on a single animal model and the incompletely elucidated mechanisms underlying the impact of FA on mitochondrial dynamics. Future investigations will explore the clinical utility of FA and its molecular mechanisms, further advancing it as a potential therapeutic for managing and delaying the progression of PD.
Collapse
Affiliation(s)
- Yina Jia
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Jing Li
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Yutong Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Yiqing Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Lei Chen
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Huaqi Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Meilan Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Hui Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| |
Collapse
|
30
|
Cai J, Chen D, Luo W, Xu F, Feng X, Zhang L, Liu H, Shen J, Ye H. The association between diverse serum folate with MAFLD and liver fibrosis based on NHANES 2017-2020. Front Nutr 2024; 11:1366843. [PMID: 38567253 PMCID: PMC10986760 DOI: 10.3389/fnut.2024.1366843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 02/27/2024] [Indexed: 04/04/2024] Open
Abstract
Background Metabolically Associated Fatty Liver Disease (MAFLD) marks a progression from the previous paradigm of Non-Alcoholic Fatty Liver Disease (NAFLD), presenting a redefined diagnostic framework that accentuates metabolic factors while recognizing non-alcoholic contributors. In our investigation, our principal aim was to scrutinize the conceivable correlation between diverse serum folate levels and the prevalence of MAFLD and liver fibrosis. Methods In our investigation, we conducted an extensive analysis utilizing data derived from the National Health and Nutrition Examination Survey (NHANES) across the years 2017-2020. We aimed to investigate the association between different serum folate concentrations and the prevalence of MAFLD and liver fibrosis by comprehensive multivariate analysis. This analytical approach considered various variables, encompassing sociodemographic characteristics, lifestyle factors, hypertension, and diabetes. By including these potential confounders in our analysis, we aimed to ensure the stability of the findings regarding the association between different serum folate concentrations and the development of MAFLD and liver fibrosis. Results In our investigation, we utilized multiple linear regression models to thoroughly analyze the data, revealing noteworthy insights. Evidently, elevated levels of both total folate and 5-MTHF exhibited a distinct negative correlation with CAP, while 5-MTHF demonstrated a notable negative correlation with LSM. Furthermore, multiple logistic regression models were employed for an in-depth examination of the data. As the concentrations of total folate and 5-MTHF in the serum increased, a substantial decrease in the likelihood of MAFLD and liver fibrosis occurrence was observed. Conclusion The findings of this investigation robustly suggest the prevalence of MAFLD and liver fibrosis decreased significantly with the increase of serum concentrations of total folate and 5-MTHF.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jianwei Shen
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Hua Ye
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
31
|
Guo P, Yu J. Association of multiple serum minerals and vitamins with metabolic dysfunction-associated fatty liver disease in US adults: National Health and Nutrition Examination Survey 2017-2018. Front Nutr 2024; 11:1335831. [PMID: 38562487 PMCID: PMC10982334 DOI: 10.3389/fnut.2024.1335831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
Background Despite the rapid increase in the global prevalence of Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD), there are no approved therapeutic drugs for MAFLD yet. Nutrient supplementation might mitigate the risk of MAFLD. It is more typical for individuals to consume multiple nutrients simultaneously. However, the studies exploring the combined effects of multiple nutrients on MAFLD are limited. This study aimed to investigate the relationship between both individual nutrients and their combined influence on the risk of MAFLD. Methods Data were obtained from National Health and Nutrition Examination Survey (NHANES), and 18 types of nutrients were considered in this study. Logistic regression analysis was performed to evaluate the correlation between single nutrients and the risk of MAFLD. The Least Absolute Shrinkage and Selection Operator (LASSO) regression analysis was performed to pinpoint the most relevant nutrient associated with the risk of MAFLD. Subsequently, both Weighted Quantile Sum (WQS) regression and Quantile g-computation (Qgcomp) were used to assess the combined effects of multiple nutrients on the risk of MAFLD. Results A total of 3,069 participants were included in this study. LASSO regression analysis showed that Se, α-tocopherol, and γ-tocopherol exhibited a positive association with the risk of MAFLD. In contrast, the serum levels of Co, P, α-cryptoxanthin, LZ, and trans-β-carotene were inversely associated with the prevalence of MAFLD. When Se and two types of vitamin E were excluded, the WQS index showed a significant inverse relationship between the remaining 15 nutrients and the risk of MAFLD; α-cryptoxanthin showed the most substantial contribution. Similarly, Qgcomp suggested that the combined effects of these 15 nutrients were associated with a lower risk of MAFLD, with α-cryptoxanthin possessing the most significant negative weights. Conclusion This study suggested that the complex nutrients with either a low proportion of Se, α-tocopherol, and γ-tocopherol or without them should be recommended for patients with MAFLD to reduce its risk.
Collapse
Affiliation(s)
| | - Jiahui Yu
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
32
|
Leclerc D, Christensen KE, Reagan AM, Keser V, Luan Y, Malysheva OV, Wasek B, Bottiglieri T, Caudill MA, Howell GR, Rozen R. Folate Deficiency and/or the Genetic Variant Mthfr 677C >T Can Drive Hepatic Fibrosis or Steatosis in Mice, in a Sex-Specific Manner. Mol Nutr Food Res 2024; 68:e2300355. [PMID: 38327171 DOI: 10.1002/mnfr.202300355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/24/2023] [Indexed: 02/09/2024]
Abstract
SCOPE Disturbances in one-carbon metabolism contribute to nonalcoholic fatty liver disease (NAFLD) which encompasses steatosis, steatohepatitis, fibrosis, and cirrhosis. The goal is to examine impact of folate deficiency and the Mthfr677C >T variant on NAFLD. METHODS AND RESULTS This study uses the new Mthfr677C >T mouse model for the human MTHFR677C >T variant. Mthfr677CC and Mthfr677TT mice were fed control diet (CD) or folate-deficient (FD) diets for 4 months. FD and Mthfr677TT alter choline/methyl metabolites in liver and/or plasma (decreased S-adenosylmethionine (SAM):S-adenosylhomocysteine (SAH) ratio, methyltetrahydrofolate, and betaine; increased homocysteine [Hcy]). FD, with contribution from Mthfr677TT, provokes fibrosis in males. Studies of normal livers reveal alterations in plasma markers and gene expression that suggest an underlying predisposition to fibrosis induced by FD and/or Mthfr677TT in males. These changes are absent or reverse in females, consistent with the sex disparity of fibrosis. Sex-based differences in methylation potential, betaine, sphingomyelin, and trimethylamine-N-oxide (TMAO) levels may prevent fibrogenesis in females. In contrast, Mthfr677TT alters choline metabolism, dysregulates expression of lipid metabolism genes, and promotes steatosis in females. CONCLUSION This study suggests that folate deficiency predisposes males to fibrosis, which is exacerbated by Mthfr677TT, whereas Mthfr677TT predisposes females to steatosis, and reveal novel contributory mechanisms for these NAFLD-related disorders.
Collapse
Affiliation(s)
- Daniel Leclerc
- Departments of Human Genetics and Pediatrics, McGill University, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Karen E Christensen
- Departments of Human Genetics and Pediatrics, McGill University, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | | | - Vafa Keser
- Departments of Human Genetics and Pediatrics, McGill University, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Yan Luan
- Departments of Human Genetics and Pediatrics, McGill University, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Olga V Malysheva
- Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY, USA
| | - Brandi Wasek
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX, USA
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX, USA
| | - Marie A Caudill
- Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY, USA
| | | | - Rima Rozen
- Departments of Human Genetics and Pediatrics, McGill University, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| |
Collapse
|
33
|
Domínguez-López I, Kovatcheva M, Casas R, Toledo E, Fitó M, Ros E, Estruch R, Serrano M, Lamuela-Raventós RM. Higher circulating vitamin B12 is associated with lower levels of inflammatory markers in individuals at high cardiovascular risk and in naturally aged mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:875-882. [PMID: 37690097 DOI: 10.1002/jsfa.12976] [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: 06/05/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Vitamin B12 is an essential nutrient that is involved in numerous physiological processes, and its deficiency can lead to various complications, including neurological and haematological disorders. Some studies have suggested that vitamin B12 may have anti-inflammatory effects, but the mechanisms underlying this relationship are not yet fully understood. We investigated the relationship between circulating vitamin B12 and inflammatory markers interleukin (IL)-6 and C-reactive protein (CRP). The association of peripheral levels of vitamin B12 with IL-6 and CRP was assessed in 136 human samples from a high cardiovascular risk population. To corroborate the results from the human trial, the analysis was replicated in naturally aged mice. RESULTS Individuals with higher serum levels of vitamin B12 showed lower concentrations of IL-6 and CRP after adjustment for potential confounders, and an inverse association was also found between serum IL-6 and vitamin B12 levels in naturally aged mice. CONCLUSION Circulating vitamin B12 was inversely associated with IL-6 and CRP in humans and with IL-6 in mice, suggesting that it may exert an anti-inflammatory effect through modulation of these pro-inflammatory molecules. © 2023 The Authors. Journal of The Science of Food and Agriculture published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Inés Domínguez-López
- Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Facultat de Farmacia, Universitat de Barcelon (UB), Barcelona, Spain
- Institut de Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Santa Coloma de Gramanet, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Kovatcheva
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Rosa Casas
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Internal Medicine, Hospital Clinic, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Estefanía Toledo
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Navarra, IdiSNA, Pamplona, Spain
| | - Montserrat Fitó
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain
| | - Emilio Ros
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Ramon Estruch
- Institut de Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Santa Coloma de Gramanet, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Internal Medicine, Hospital Clinic, Institut d'Investigació Biomèdica August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Cambridge Institute of Science, Altos Labs, Cambridge, UK
| | - Rosa M Lamuela-Raventós
- Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Facultat de Farmacia, Universitat de Barcelon (UB), Barcelona, Spain
- Institut de Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Santa Coloma de Gramanet, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
34
|
Liu K, Chen Y, Chen J, Chen W, Sun X, Mao Y, Ye D. Genetically determined circulating micronutrients and the risk of nonalcoholic fatty liver disease. Sci Rep 2024; 14:1105. [PMID: 38212362 PMCID: PMC10784479 DOI: 10.1038/s41598-024-51609-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/07/2024] [Indexed: 01/13/2024] Open
Abstract
Evidence from epidemiological literature on the association of circulating micronutrients with risk of nonalcoholic fatty liver disease (NAFLD) is inconsistent. We aimed to elucidate the causal relationships using Mendelian randomization (MR). Single-nucleotide polymorphisms associated with 14 circulating micronutrients (β-carotene, calcium, copper, folate, iron, magnesium, phosphorus, selenium, vitamin B6, B12, C, D, K1 and zinc) were employed as instrumental variables. Summary level data for NAFLD were obtained from a genome-wide association study (GWAS) meta-analysis of 8434 cases and 770,180 controls (discovery stage) and another two datasets including 1483 NAFLD cases and 17,781 controls (replication stage 1) and 2134 NAFLD cases and 33,433 controls (replication stage 2). Inverse variance-weighted method (IVW) was used as primary analysis, supplemented with a series of sensitivity analysis. Genetically predicted higher β‑carotene levels were suggestively associated with reduced NAFLD risk [odds ratio (OR) 0.81, 95% confidence interval (CI) 0.66-0.99; P = 0.047], whereas the association did not survive the false discovery rates (FDR) correction (PFDR = 0.164). Genetically predicted circulating iron (OR 1.16, 95% CI 1.05-1.29; P = 0.006, PFDR = 0.028), selenium (OR 1.11, 95% CI 1.03-1.20; P = 0.005, PFDR = 0.028) and vitamin B12 (OR 1.08, 95% CI 1.03-1.13; P = 0.002, PFDR = 0.028) were significantly associated with increased risk of NAFLD. Moreover, the findings were consistent in individual datasets (Pheterogeneity > 0.05) and confirmed in sensitivity analysis. Our study provided evidence that circulating iron, selenium and vitamin B12 might be causally linked to the risk of NAFLD, which deserves further exploration of the potential biological mechanism.
Collapse
Affiliation(s)
- Ke Liu
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Ying Chen
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Jiaxin Chen
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Weiwei Chen
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Xiaohui Sun
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Yingying Mao
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Ding Ye
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
35
|
Yang Y, Ke Y, Liu X, Zhang Z, Zhang R, Tian F, Zhi L, Zhao G, Lv B, Hua S, Wu H. Navigating the B vitamins: Dietary diversity, microbial synthesis, and human health. Cell Host Microbe 2024; 32:12-18. [PMID: 38211561 DOI: 10.1016/j.chom.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/09/2023] [Accepted: 12/12/2023] [Indexed: 01/13/2024]
Abstract
B vitamins are intricately involved in various physiological processes vital for health. Their significance is complicated by the heterogeneous landscape of B vitamin distribution in diets and the contributions of the gut microbiota. Here, we delve into the impact of these factors on B vitamins and introduce strategies, with a focus on microbiota-based therapeutic options, to enhance their availability for improved well-being. Additionally, we provide an ecological and evolutionary perspective on the importance of B vitamins to human-microbiota interactions. In the dynamic realms of nutrition and microbiome science, these essential micronutrients continue to play a fundamental role in our understanding of disease development.
Collapse
Affiliation(s)
- Yudie Yang
- State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Yize Ke
- State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Xinyan Liu
- State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Zhidong Zhang
- State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200433, China; College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Rongji Zhang
- State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200433, China; College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Fang Tian
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Luqian Zhi
- State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Guoping Zhao
- State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Bomin Lv
- State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200433, China.
| | - Sha Hua
- Department of Cardiovascular Medicine, Heart Failure Center, Ruijin Hospital Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Hao Wu
- State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200433, China; Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
36
|
Tripathi M, Gauthier K, Sandireddy R, Zhou J, Gupta P, Sakthivel S, Jiemin N, Arul K, Tikno K, Park SH, Wang L, Ho L, Giguere V, Ghosh S, McDonnell DP, Yen PM, Singh BK. Estrogen receptor-related receptor (Esrra) induces ribosomal protein Rplp1-mediated adaptive hepatic translation during prolonged starvation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574937. [PMID: 38260502 PMCID: PMC10802477 DOI: 10.1101/2024.01.09.574937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Protein translation is an energy-intensive ribosome-driven process that is reduced during nutrient scarcity to conserve cellular resources. During prolonged starvation, cells selectively translate specific proteins to enhance their survival (adaptive translation); however, this process is poorly understood. Accordingly, we analyzed protein translation and mRNA transcription by multiple methods in vitro and in vivo to investigate adaptive hepatic translation during starvation. While acute starvation suppressed protein translation in general, proteomic analysis showed that prolonged starvation selectively induced translation of lysosome and autolysosome proteins. Significantly, the expression of the orphan nuclear receptor, estrogen-related receptor alpha (Esrra) increased during prolonged starvation and served as a master regulator of this adaptive translation by transcriptionally stimulating 60S acidic ribosomal protein P1 (Rplp1) gene expression. Overexpression or siRNA knockdown of Esrra expression in vitro or in vivo led to parallel changes in Rplp1 gene expression, lysosome/autophagy protein translation, and autophagy. Remarkably, we have found that Esrra had dual functions by not only regulating transcription but also controling adaptive translation via the Esrra/Rplp1/lysosome/autophagy pathway during prolonged starvation.
Collapse
Affiliation(s)
- Madhulika Tripathi
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Karine Gauthier
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46 Allée d’Italie, 69364 Lyon Cedex 07, France
| | - Reddemma Sandireddy
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Jin Zhou
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Priyanka Gupta
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Suganya Sakthivel
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Nah Jiemin
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, Québec H3A 1A3, Canada
| | - Kabilesh Arul
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Keziah Tikno
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Sung-Hee Park
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C238A Levine Science Research Center, Durham, NC 27710, USA
| | - Lijin Wang
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Lena Ho
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Vincent Giguere
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, Québec H3A 1A3, Canada
| | - Sujoy Ghosh
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Donald P. McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C238A Levine Science Research Center, Durham, NC 27710, USA
| | - Paul M. Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
- Duke Molecular Physiology Institute and Dept. of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Brijesh K. Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| |
Collapse
|
37
|
Raza S, Rajak S, Singh R, Zhou J, Sinha RA, Goel A. Cell-type specific role of autophagy in the liver and its implications in non-alcoholic fatty liver disease. World J Hepatol 2023; 15:1272-1283. [PMID: 38192406 PMCID: PMC7615497 DOI: 10.4254/wjh.v15.i12.1272] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/07/2023] [Accepted: 12/08/2023] [Indexed: 12/25/2023] Open
Abstract
Autophagy, a cellular degradative process, has emerged as a key regulator of cellular energy production and stress mitigation. Dysregulated autophagy is a common phenomenon observed in several human diseases, and its restoration offers curative advantage. Non-alcoholic fatty liver disease (NAFLD), more recently renamed metabolic dysfunction-associated steatotic liver disease, is a major metabolic liver disease affecting almost 30% of the world population. Unfortunately, NAFLD has no pharmacological therapies available to date. Autophagy regulates several hepatic processes including lipid metabolism, inflammation, cellular integrity and cellular plasticity in both parenchymal (hepatocytes) and non-parenchymal cells (Kupffer cells, hepatic stellate cells and sinusoidal endothelial cells) with a profound impact on NAFLD progression. Understanding cell type-specific autophagy in the liver is essential in order to develop targeted treatments for liver diseases such as NAFLD. Modulating autophagy in specific cell types can have varying effects on liver function and pathology, making it a promising area of research for liver-related disorders. This review aims to summarize our present understanding of cell-type specific effects of autophagy and their implications in developing autophagy centric therapies for NAFLD.
Collapse
Affiliation(s)
- Sana Raza
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Uttar Pradesh, Lucknow 226014, India
| | - Sangam Rajak
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Uttar Pradesh, Lucknow 226014, India
| | - Rajani Singh
- Department of Hepatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Uttar Pradesh, Lucknow 226014, India
| | - Jin Zhou
- CVMD, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Rohit A Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Uttar Pradesh, Lucknow 226014, India
| | - Amit Goel
- Department of Hepatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Uttar Pradesh, Lucknow 226014, India.
| |
Collapse
|
38
|
Wu JY, Liu MY, Hung KC, Hsu WH, Tsai YW, Liu TH, Huang PY, Chuang MH, Hsieh YL, Lai CC, Kuo YH. Nutritional deficiency anemia and post-acute sequelae in patients with severe acute respiratory syndrome coronavirus 2 infection: A six-month retrospective cohort analysis of 30 892 patients. J Med Virol 2023; 95:e29246. [PMID: 38010833 DOI: 10.1002/jmv.29246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/14/2023] [Accepted: 11/07/2023] [Indexed: 11/29/2023]
Abstract
The effect of anemia on the post-acute outcome of patients with severe acute respiratory syndrome coronavirus 2 infection was unclear. This study aimed to investigate the potential association between nutritional deficiency anemia (NDA) status and post-acute sequelae of patients with SARS-CoV-2 infection. This retrospective cohort study included patients with coronavirus disease (COVID-19) from January 1, 2022 to November 30, 2022 using the TriNetX research network. The patients were grouped into the NDA group comprising patients diagnosed with NDA and the control group comprising patients without NDA, and propensity score matching (PSM) was performed to balance the two groups. The primary outcome was a composite of post-COVID-19 condition, all-cause hospitalization, and all-cause death. The secondary outcomes were any individual outcomes of the primary composite. The follow-up period was set at 90-180 days after COVID-19 diagnosis. Two cohorts comprising 15 446 nonhospitalized patients with COVID-19 in each group with balanced baseline characteristics were created using PSM. During the follow-up period, the NDA group demonstrated a higher risk of the composite primary outcome, including post-COVID-19 condition, all-cause hospitalization, or all-cause death (hazard ratio [HR], 1.896; 95% confidence interval [CI] = 1.757-2.045). Regarding secondary outcomes, the NDA group was associated with worse outcomes, including post-COVID-19 condition (HR, 1.992; 95% CI = 1.403-2.828), all-cause hospitalization (HR, 1.856; 95% CI = 1.714-2.009), and all-cause death (HR, 3.922; 95% CI = 2.910-5.285) compared to the control group. Among nonhospitalized patients with COVID-19, NDA was associated with a higher risk of post-COVID-19 condition, all-cause hospitalization, and all-cause death during the 90-180-day follow-up period.
Collapse
Affiliation(s)
- Jheng-Yan Wu
- Department of Nutrition, Chi Mei Medical Center, Tainan, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Mei-Yuan Liu
- Department of Nutrition, Chi Mei Medical Center, Tainan, Taiwan
- Department of Nutrition and Health Sciences, Chang Jung Christian University, Tainan, Taiwan
- Department of Food Nutrition, Chung Hwa University of Medical Technology, Tainan, Taiwan
- Department of Health and Nutrition, Chia Nan University of Pharmacy & Science, Tainan, Taiwan
| | - Kuo-Chuan Hung
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - Wan-Hsuan Hsu
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Ya-Wen Tsai
- Center of Integrative Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Ting-Hui Liu
- Department of Psychiatry, Chi Mei Medical Center, Tainan, Taiwan
| | - Po-Yu Huang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Min-Hsiang Chuang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Ya-Ling Hsieh
- Department of Nursing, Chi Mei Medical Center, Tainan, Taiwan
- Graduate Institute of Nursing, College of Health Care, China Medical University, Taichung, Taiwan
| | - Chi-Cheng Lai
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung City, Taiwan
- Department of Internal Medicine, Division of Hospital Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Yu-Hsuan Kuo
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
- College of Pharmacy and Science, Chia Nan University, Tainan, Taiwan
| |
Collapse
|
39
|
Zhang Z, Lu W, Liu P, Li M, Ge X, Yu B, Wu Z, Liu G, Ding N, Cui B, Chen X. Microbial modifications with Lycium barbarum L. oligosaccharides decrease hepatic fibrosis and mitochondrial abnormalities in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155068. [PMID: 37690228 DOI: 10.1016/j.phymed.2023.155068] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/03/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Lycium barbarum L. is a typical Chinese herbal and edible plant and are now consumed globally. Low molecular weight L. barbarum L. oligosaccharides (LBO) exhibit better antioxidant activity and gastrointestinal digestibility in vitro than high molecular weight polysaccharides. However, the LBO on the treatment of liver disease is not studied. PURPOSE Modification of the gut microbial ecosystem by LBO is a promising treatment for liver fibrosis. STUDY DESIGN AND METHODS Herein, LBO were prepared and characterized. CCl4-treated mice were orally gavaged with LBO and the effects on hepatic fibrosis and mitochondrial abnormalities were evaluated according to relevant indicators (gut microbiota, faecal metabolites, and physiological and biochemical indices). RESULTS The results revealed that LBO, a potential prebiotic source, is a pyranose cyclic oligosaccharide possessing α-glycosidic and β-glycosidic bonds. Moreover, LBO supplementation restored the configuration of the bacterial community, enhanced the proliferation of beneficial species in the gastrointestinal tract (e.g., Bacillus, Tyzzerella, Fournierella and Coriobacteriaceae UCG-002), improved microbial metabolic alterations (i.e., carbohydrate metabolism, vitamin metabolism and entero-hepatic circulation), and increased antioxidants, including doxepin, in mice. Finally, LBO administration reduced serum inflammatory cytokine and hepatic hydroxyproline levels, improved intestinal and hepatic mitochondrial functions, and ameliorated mouse liver fibrosis. CONCLUSION These findings indicate that LBO can be utilized as a prebiotic and has a remarkable ability to mitigate liver fibrosis.
Collapse
Affiliation(s)
- Zheng Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Wenjia Lu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Pengfei Liu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Mengjie Li
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Xinyi Ge
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Bin Yu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Zhengzong Wu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Guimei Liu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Nannan Ding
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Bo Cui
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China.
| | - Xiao Chen
- College of Health Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250353, China.
| |
Collapse
|
40
|
Li J, Huang J, Lv Y, Ji H. Association between dietary intakes of B vitamins and nonalcoholic fatty liver disease in postmenopausal women: a cross-sectional study. Front Nutr 2023; 10:1272321. [PMID: 37927496 PMCID: PMC10621796 DOI: 10.3389/fnut.2023.1272321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is increasingly common globally, particularly among postmenopausal women. Diet plays a fundamental role in the treatment of NAFLD. However, clinical research on the dietary intakes of B vitamins, specifically in postmenopausal women, is scant. Hence, it is imperative to study the impact of B vitamin dietary intake in postmenopausal women. Methods This study utilized National Health and Nutrition Examination Survey (NHANES) data for 668 postmenopausal women. Logistic regression analysis was conducted to investigate the association of the intakes of B vitamins with hepatic steatosis and liver fibrosis prevalence. The analysis accounted for various covariates and employed restricted cubic spline analysis to examine potential nonlinear relationships. Additionally, interactions among age, diabetes, and B-vitamin intakes, as well as the interaction between folate and vitamin B12 intake, were explored. Results Higher intakes of folate [0.30 (0.10-0.88)], choline [0.26 (0.07-0.95)], vitamin B1, and vitamin B2 were associated with a reduced risk of hepatic steatosis in postmenopausal women. The associations of niacin (P-nonlinear = 0.0003), vitamin B1 (P-nonlinear = 0.036), and vitamin B2 (P-nonlinear<0.0001) intakes with hepatic steatosis showed a nonlinear pattern. However, no significant associations were observed between the intakes of niacin, vitamin B6 and vitamin B12 and hepatic steatosis. Furthermore, there were no significant associations between B-vitamin intakes and liver fibrosis. No interaction effects were observed. Conclusion Dietary intakes of folate, choline, vitamin B1, and vitamin B2 may be associated with liver steatosis in postmenopausal women, these results suggest that optimizing the intake of these specific B vitamins may have a protective effect against liver steatosis in postmenopausal women, offering valuable insights into potential dietary strategies to promote their well-being.
Collapse
Affiliation(s)
- Jiajie Li
- Department of Hepatobiliary and Pancreatic Medicine, Infectious Disease. and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jingda Huang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yanqing Lv
- Department of Hepatobiliary and Pancreatic Medicine, Infectious Disease. and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Huifan Ji
- Department of Hepatobiliary and Pancreatic Medicine, Infectious Disease. and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
41
|
Schwenger KJP, Ghorbani Y, Rezaei K, Fischer SE, Jackson TD, Okrainec A, Allard JP. Relationship between dietary intake components and hepatic fibrosis in those with obesity before and 1 year after bariatric surgery. Nutrition 2023; 114:112095. [PMID: 37437418 DOI: 10.1016/j.nut.2023.112095] [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: 02/24/2023] [Revised: 05/05/2023] [Accepted: 05/22/2023] [Indexed: 07/14/2023]
Abstract
OBJECTIVES Non-alcoholic fatty liver disease is highly prevalent in the bariatric population but not all patients develop liver fibrosis. Considering that fibrosis may affect clinical outcomes, it is important to assess and treat contributing factors. In this population, it is not clear whether dietary intake is a contributor. The objective was to determine the relationship between dietary intake components and liver fibrosis before and 1 y after Roux-en-Y gastric bypass (RYGB). METHODS This was a prospective cross-sectional (n = 133) study conducted between 2013 and 2022. In addition, a subgroup of 44 patients were followed for 1 y post-RYGB. Anthropometrics, biochemical measurements, and 3-d food records and liver biopsies were obtained presurgery and, in a subgroup of patients, as for the cohort, 1 y post-RYGB. RESULTS In the cross-sectional study, 78.2% were female, with a median age of 48 y and body mass index of 46.8 kg/m2; 33.8% had type 2 diabetes mellitus and 57.1% had metabolic syndrome. In a multivariate analysis, age (odds ratio; 95% CI) (1.076; 1.014-1.141), alanine transaminase (1.068; 1.025-1.112), calorie intake (1.001; 1.000-1.002), and dietary copper (0.127; 0.022-0.752) were independently associated with fibrosis (<0.05). At 1 y post-RYGB, no independent risk factors were associated with persistent fibrosis. CONCLUSIONS In bariatric patients before surgery, higher age, alanine transaminase, and total calorie and lower copper intakes were independent risk factors associated with liver fibrosis. These relationships were no longer observed after RYGB, likely due to the effect of surgery on weight and similar postsurgery diet among patients.
Collapse
Affiliation(s)
| | - Yasaman Ghorbani
- Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Kuorosh Rezaei
- Toronto General Hospital, University Health Network, Toronto, Canada
| | - Sandra E Fischer
- Toronto General Hospital, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Timothy D Jackson
- Division of Surgery, University of Toronto, Toronto, Ontario, Canada; Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Allan Okrainec
- Division of Surgery, University of Toronto, Toronto, Ontario, Canada; Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Johane P Allard
- Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
42
|
Kovalska M, Hnilicova P, Kalenska D, Adamkov M, Kovalska L, Lehotsky J. Alzheimer's Disease-like Pathological Features in the Dorsal Hippocampus of Wild-Type Rats Subjected to Methionine-Diet-Evoked Mild Hyperhomocysteinaemia. Cells 2023; 12:2087. [PMID: 37626897 PMCID: PMC10453870 DOI: 10.3390/cells12162087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/27/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Multifactorial interactions, including nutritional state, likely participate in neurodegeneration's pathogenesis and evolution. Dysregulation in methionine (Met) metabolism could lead to the development of hyperhomocysteinaemia (hHcy), playing an important role in neuronal dysfunction, which could potentially lead to the development of Alzheimer's disease (AD)-like pathological features. This study combines proton magnetic resonance spectroscopy (1H MRS) with immunohistochemical analysis to examine changes in the metabolic ratio and histomorphological alterations in the dorsal rat hippocampus (dentate gyrus-DG) subjected to a high Met diet. Male Wistar rats (420-480 g) underwent hHcy evoked by a Met-enriched diet (2 g/kg of weight/day) lasting four weeks. Changes in the metabolic ratio profile and significant histomorphological alterations have been found in the DG of hHcy rats. We have detected increased morphologically changed neurons and glial cells with increased neurogenic markers and apolipoprotein E positivity parallel with a diminished immunosignal for the N-Methyl-D-Aspartate receptor 1 in hHcy animals. A Met diet induced hHcy, likely via direct Hcy neurotoxicity, an interference with one carbon unit metabolism, and/or epigenetic regulation. These conditions lead to the progression of neurodegeneration and the promotion of AD-like pathological features in the less vulnerable hippocampal DG, which presents a plausible therapeutic target.
Collapse
Affiliation(s)
- Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.K.); (M.A.)
| | - Petra Hnilicova
- Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Marian Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.K.); (M.A.)
| | - Libusa Kovalska
- Clinic of Stomatology and Maxillofacial Surgery, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Jan Lehotsky
- Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
43
|
Chen HK, Luo J, Li XJ, Liao WZ, Hu YQ, Guo XG. Serum folate associated with nonalcoholic fatty liver disease and advanced hepatic fibrosis. Sci Rep 2023; 13:12933. [PMID: 37558738 PMCID: PMC10412549 DOI: 10.1038/s41598-023-39641-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023] Open
Abstract
The role played by serum folate in the progression of nonalcoholic fatty liver disease (NAFLD) remains controversial. The purpose of this study was to investigate the association of serum folate with NAFLD and advanced liver fibrosis (AHF). We conducted a cross-sectional study with 5417 participants using 2011-2018 NHANES data. Multiple logistic regression analysis and propensity score matching analysis were used to investigate the association of serum folate with NAFLD and AHF. In the completely adjusted model, participants in the high serum folate group had a 27% (OR 0.73, 95% CI 0.62, 0.87, p = 0.0003) and 53% (OR 0.47, 95% CI 0.35, 0.63, p < 0.0001) lower odds of suffering from NAFLD and AHF, respectively, compared to the low serum folate group. The similar results in propensity score matching further validated the above association. Stratified analysis showed that the negative correlation of serum folate with NAFLD and AHF demonstrated a broad consistency across populations. The results of this study indicate that higher serum folate level was associated with lower odds of NAFLD and AHF among US adults. Further prospective studies are necessary due to the limitations of cross-sectional studies.
Collapse
Affiliation(s)
- Hao-Kai Chen
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Jing Luo
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Xiu-Juan Li
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Wan-Zhe Liao
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Nanshan College of Guangzhou Medical University, Guangzhou, China
| | - Yu-Qi Hu
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Xu-Guang Guo
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
44
|
Wu C, Duan X, Wang X, Wang L. Advances in the role of epigenetics in homocysteine-related diseases. Epigenomics 2023; 15:769-795. [PMID: 37718931 DOI: 10.2217/epi-2023-0207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023] Open
Abstract
Homocysteine has a wide range of biological effects. However, the specific molecular mechanism of its pathogenicity is still unclear. The diseases induced by hyperhomocysteinemia (HHcy) are called homocysteine-related diseases. Clinical treatment of HHcy is mainly through folic acid and B-complex vitamins, which are not effective in reducing the associated end point events. Epigenetics is the alteration of heritable genes caused by DNA methylation, histone modification, noncoding RNAs and chromatin remodeling without altering the DNA sequence. In recent years the role of epigenetics in homocysteine-associated diseases has been gradually discovered. This article summarizes the latest evidence on the role of epigenetics in HHcy, providing new directions for its prevention and treatment.
Collapse
Affiliation(s)
- Chengyan Wu
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Xulei Duan
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Xuehui Wang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Libo Wang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
45
|
Zhang Y, Ouyang J, Zhan L, Li Y, Li S, He Y, Wang H, Zhang X. Autophagy in homocysteine‑induced HUVEC senescence. Exp Ther Med 2023; 26:354. [PMID: 37324509 PMCID: PMC10265697 DOI: 10.3892/etm.2023.12053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 04/27/2023] [Indexed: 06/17/2023] Open
Abstract
The senescence of vascular endothelial cells (VECs) drives the occurrence and development of cardiovascular disease (CVD). Homocysteine (HCY) is a general risk factor for age-associated CVDs. Autophagy, an evolutionarily conserved lysosomal protein degradation pathway, serves a part in VEC senescence. The purpose of this study was to investigate the role of autophagy in HCY-induced endothelial cell senescence and explore novel mechanisms and therapeutic approaches for related CVDs. Human umbilical vein endothelial cells (HUVECs) were isolated from fresh umbilical cords of healthy pregnancies. Cell Counting Kit-8, flow cytometry and senescence-associated (SA) β-galactosidase (Gal) staining demonstrated that HCY induced HUVEC senescence by decreasing cell proliferation, arresting cell cycle and increasing the number of SA-β-Gal-positive cells. Stub-RFP-Sens-GFP-LC3 autophagy-related double fluorescence lentivirus revealed that HCY increased autophagic flux. Further, inhibition of autophagy using 3-methyladenine increased HCY-induced HUVEC senescence. By contrast, the induction of autophagy via rapamycin alleviated HCY-induced HUVEC senescence. Finally, the detection of reactive oxygen species (ROS) with ROS kit showed that HCY increased intracellular ROS, whereas induction of autophagy reduced intracellular ROS. In conclusion, HCY increased HUVEC senescence and upregulated autophagy; moderate autophagy could reverse HCY-induced cell senescence. Autophagy may alleviate HCY-induced cell senescence by decreasing intracellular ROS. This provides insight into the underlying mechanism of HCY-induced VEC senescence and potential treatments for age-associated CVDs.
Collapse
Affiliation(s)
- Yexi Zhang
- Department of Functional Examination, Northern Jiangsu People's Hospital, The Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu 225000, P.R. China
| | - Juyan Ouyang
- Department of Geriatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumchi, Xinjiang Uyghur Autonomous Region 830000, P.R. China
| | - Liu Zhan
- Department of Functional Science, Karamay College of Xinjiang Medical University, Karamay, Xinjiang Uyghur Autonomous Region 834000, P.R. China
| | - Yu Li
- Department of Geriatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumchi, Xinjiang Uyghur Autonomous Region 830000, P.R. China
| | - Shaoshan Li
- Department of Clinical Teaching, Karamay College of Xinjiang Medical University, Karamay, Xinjiang Uyghur Autonomous Region 834000, P.R. China
| | - Yi He
- Department of Morphology, Karamay College of Xinjiang Medical University, Karamay, Xinjiang Uyghur Autonomous Region 834000, P.R. China
| | - Hong Wang
- Department of Geriatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumchi, Xinjiang Uyghur Autonomous Region 830000, P.R. China
| | - Xiangyang Zhang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumchi, Xinjiang Uyghur Autonomous Region 830000, P.R. China
| |
Collapse
|
46
|
Baradeiya AM, Taghlabi KM, Saleh AN, Manikonda S, Salim SS. Can Nutritional Supplements Benefit Patients With Nonalcoholic Steatohepatitis and Nonalcoholic Fatty Liver Disease? Cureus 2023; 15:e40849. [PMID: 37489221 PMCID: PMC10363331 DOI: 10.7759/cureus.40849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2023] [Indexed: 07/26/2023] Open
Abstract
A characteristic of nonalcoholic fatty liver disease (NAFLD) is the buildup of excess fat in the liver which encompasses various clinical phases, including steatosis, inflammation, ballooning, fibrosis, and liver cirrhosis. Nonalcoholic steatohepatitis (NASH) represents a severe form of NAFLD. The prevalence of NAFLD, particularly NASH, is notably high among Hispanics and those with morbid obesity. Diabetes, obesity, and dyslipidemia are significant risk factors in patients with NAFLD. The pathogenesis of NAFLD involves complex interactions between hormonal, nutritional, and genetic factors. Different clinical trials have been conducted to determine if there are any supplements that could help patients with NASH. Evidence has shown that vitamin E decreased the NAFLD activity score but not fibrosis. Our review summarizes the influence of supplementation on patients with NAFLD and NASH, focusing on the use of different clinical trials, systematic reviews, and meta-analyses. In the future, patients and physicians will play crucial roles in exploring diverse approaches and finding effective solutions to address this growing issue.
Collapse
Affiliation(s)
- Ahmed M Baradeiya
- Advanced Liver Therapies Research, Baylor College of Medicine, Houston, USA
| | | | | | | | - Siffat S Salim
- Surgery, Holy Family Red Crescent Medical College Hospital, Dhaka, BGD
| |
Collapse
|
47
|
Molaqanbari MR, Zarringol S, Talari HR, Taghizadeh M, Bahmani F, Mohtashamian A, Ebrahimzadeh A, Sharifi N. Effects of Folic Acid Supplementation on Liver Enzymes, Lipid Profile, and Insulin Resistance in Patients with Non-Alcoholic Fatty Liver Disease: A Randomized Controlled Trial. Adv Biomed Res 2023; 12:103. [PMID: 37288023 PMCID: PMC10241628 DOI: 10.4103/abr.abr_90_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 06/09/2023] Open
Abstract
Background Previous evidence revealed an association between folate deficiency and non-alcoholic fatty liver disease (NAFLD). This study is the first one investigating the effects of folic acid on hepatic steatosis grade, liver enzymes, insulin resistance, and lipid profile in NAFLD cases. Materials and Methods Sixty-six participants with NAFLD were allocated randomly to take either a placebo or one oral tablet of folic acid (1 mg) on a daily basis within eight weeks. Serum folate, homocysteine, glucose, aminotransferases, insulin, homeostasis model assessment of insulin resistance (HOMA-IR), and lipids were assessed. Ultrasonography was used for assessing the liver steatosis grade. Results The serum alanine transaminase, grade of hepatic steatosis, and aspartate transaminase significantly were decreased within both study groups; however, the between-group comparison was not statistically significant. Of note, the decrease in ALT was more pronounced in folic acid compared with the placebo group (-5.45 ± 7.45 vs. -2.19 ± 8.6 IU/L). The serum homocysteine was decreased after receiving folic acid compared to the placebo (-0.58 ± 3.41 vs. +0.4 ± 3.56 μmol/L; adjusted P = 0.054). Other outcomes did not significantly change. Conclusion Supplementation with folic acid (1 mg/d) for eight weeks among cases with NAFLD did not change significantly the serum levels of liver enzymes, the hepatic steatosis grade, insulin resistance and lipid profile. However, it was able to prevent the increase in homocysteine in comparison with the placebo. Conducting further research is suggested with the longer duration and different doses of folic acid, adjusted to the genotypes of methylenetetrahydrofolate reductase polymorphism, among NAFLD patients.
Collapse
Affiliation(s)
- Mohamad Reza Molaqanbari
- Department of Internal Medicine, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Shadi Zarringol
- Department of Internal Medicine, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamid Reza Talari
- Department of Radiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Basic Science Research Institute, Kashan University of Medical Sciences, Kashan, Iran
| | - Fereshteh Bahmani
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Basic Science Research Institute, Kashan University of Medical Sciences, Kashan, Iran
| | - Abbas Mohtashamian
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Basic Science Research Institute, Kashan University of Medical Sciences, Kashan, Iran
| | - Armin Ebrahimzadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Basic Science Research Institute, Kashan University of Medical Sciences, Kashan, Iran
| | - Nasrin Sharifi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Basic Science Research Institute, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
48
|
Sinha RA. Autophagy: A Cellular Guardian against Hepatic Lipotoxicity. Genes (Basel) 2023; 14:553. [PMID: 36874473 PMCID: PMC7614268 DOI: 10.3390/genes14030553] [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] [Indexed: 02/25/2023] Open
Abstract
Lipotoxicity is a phenomenon of lipid-induced cellular injury in nonadipose tissue. Excess of free saturated fatty acids (SFAs) contributes to hepatic injury in nonalcoholic fatty liver disease (NAFLD), which has been growing at an unprecedented rate in recent years. SFAs and their derivatives such as ceramides and membrane phospholipids have been shown to induce intrahepatic oxidative damage and ER stress. Autophagy represents a cellular housekeeping mechanism to counter the perturbation in organelle function and activation of stress signals within the cell. Several aspects of autophagy, including lipid droplet assembly, lipophagy, mitophagy, redox signaling and ER-phagy, play a critical role in mounting a strong defense against lipotoxic lipid species within the hepatic cells. This review provides a succinct overview of our current understanding of autophagy-lipotoxicity interaction and its pharmacological and nonpharmacological modulation in treating NAFLD.
Collapse
Affiliation(s)
- Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| |
Collapse
|
49
|
Reply to: "Hyperhomocysteinemia predicts liver-related clinical outcomes in the general population". J Hepatol 2023; 78:e174-e175. [PMID: 36736736 DOI: 10.1016/j.jhep.2023.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 02/05/2023]
|
50
|
Abstract
Methotrexate is a key component of the treatment of inflammatory rheumatic diseases and the mainstay of therapy in rheumatoid arthritis. Hepatotoxicity has long been a concern for prescribers envisaging long-term treatment with methotrexate for their patients. However, the putative liver toxicity of methotrexate should be evaluated in the context of advances in our knowledge of the pathogenesis and natural history of liver disease, especially non-alcoholic fatty liver disease (NAFLD). Notably, patients with NAFLD are at increased risk for methotrexate hepatotoxicity, and methotrexate can worsen the course of NAFLD. Understanding the mechanisms of acute hepatotoxicity can facilitate the interpretation of elevated concentrations of liver enzymes in this context. Liver fibrosis and the mechanisms of fibrogenesis also need to be considered in relation to chronic exposure to methotrexate. A number of non-invasive tests for liver fibrosis are available for use in patients with rheumatic disease, in addition to liver biopsy, which can be appropriate for particular individuals. On the basis of the available evidence, practical suggestions for pretreatment screening and long-term monitoring of methotrexate therapy can be made for patients who have (or are at risk for) chronic liver disease.
Collapse
|