1
|
Zarandi PK, Ghiasi M, Heiat M. The role and function of lncRNA in ageing-associated liver diseases. RNA Biol 2025; 22:1-8. [PMID: 39697114 PMCID: PMC11660375 DOI: 10.1080/15476286.2024.2440678] [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] [Revised: 10/09/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Liver diseases are a significant global health issue, characterized by elevated levels of disorder and death. The substantial impact of ageing on liver diseases and their prognosis is evident. Multiple processes are involved in the ageing process, which ultimately leads to functional deterioration of this organ. The process of liver ageing not only renders the liver more susceptible to diseases but also compromises the integrity of other organs due to the liver's critical function in metabolism regulation. A growing body of research suggests that long non-coding RNAs (lncRNAs) play a significant role in the majority of pathophysiological pathways. They regulate gene expression through a variety of interactions with microRNAs (miRNAs), messenger RNAs (mRNAs), DNA, or proteins. LncRNAs exert a major influence on the progression of age-related liver diseases through the regulation of cell proliferation, necrosis, apoptosis, senescence, and metabolic reprogramming. A concise overview of the current understanding of lncRNAs and their potential impact on the development of age-related liver diseases will be provided in this mini-review.
Collapse
Affiliation(s)
- Peyman Kheirandish Zarandi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Cancer Biology Signaling Pathway Interest Group (CBSPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohsen Ghiasi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Xie J, Li B, He C, Yang S, Yang S, Sun P, Zhu G, Wang J. The mechanism underlying of Zuoguiyin on liver and kidney in D-gal-induced subacute aging female rats: A perspective on SIRT1-PPARγ pathway regulation of oxidative stress, inflammation and apoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119811. [PMID: 40239881 DOI: 10.1016/j.jep.2025.119811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 04/01/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aging is a complex biological metabolic process. Traditional Chinese Medicine (TCM) theory posits that deficiencies in the liver and kidneys in women is closely associated with aging. Zuoguiyin (ZGY), originating from the Jing Yue Quan Shu (A.D. 1624), is renowned for its efficacy in nourishing Yin and tonifying the kidneys. Pharmacological studies have confirmed that ZGY could enhance liver and kidney functions in aging females, but its molecular mechanisms remain unclear. AIM OF THE STUDY This study aims to investigate the effects and potential mechanisms of ZGY on the liver and kidneys in female subacute aging model rats. MATERIALS AND METHODS A combination of network pharmacology and animal experiments was employed to identify the active components and potential targets of ZGY. A subacute aging model was established in female rats using D-galactose (D-gal) injection. After intervention with ZGY, its effects and mechanisms on the liver and kidney were evaluated using behavioral tests, ELISA, H&E staining, immunohistochemistry, Western blot analysis, and molecular docking. RESULTS Network pharmacology analysis indicated that the effects of ZGY on subacute aging female rats might be associated with the regulation of oxidative stress and inflammatory responses via the sirtuin 1 (SIRT1)- peroxisome proliferator-activated receptor gamma (PPARγ) pathway. Compared to female subacute aging model rats, ZGY significantly improved behaviors in model rats. It notably reduced levels of MDA, NF-κB, and IL-6 in serum, liver, and kidney tissues, while increasing the levels of SOD, CAT, and IL-10. Furthermore, ZGY enhanced liver and kidney functions, alleviated pathological damage to these organs, decreased BAX expression, increased Bcl-2 expression, and inhibited apoptosis in liver and kidney cells. Western blot analysis further validated that ZGY activates the expression of proteins in the SIRT1-PPARγ pathway. Molecular docking implicated that 65 enter blood components of ZGY might be the active components. CONCLUSION This study reveals that ZGY exerts positive effects on the liver and kidney in D-gal-induced subacute aging female rats. Its mechanisms are associated with the activation of the SIRT1-PPARγ pathway, which regulates oxidative stress, inflammation, and apoptotic pathways.
Collapse
Affiliation(s)
- Jingru Xie
- The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230061, China.
| | - Baobao Li
- Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Cunbao He
- Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Shubin Yang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Shaojie Yang
- The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230061, China.
| | - Peiyang Sun
- The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230061, China.
| | - Guoqi Zhu
- Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Jingji Wang
- The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230061, China.
| |
Collapse
|
3
|
Chaudhary JK, Danga AK, Kumari A, Bhardwaj A, Rath PC. Role of Stem Cells in Ageing and Age-related Diseases. Mech Ageing Dev 2025:112069. [PMID: 40324541 DOI: 10.1016/j.mad.2025.112069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Stem cell functions and ageing are deeply interconnected, continually influencing each other in multiple ways. Stem cells play a vital role in organ maintenance, regeneration, and homeostasis, all of which decline over time due to gradual reduction in their self-renewal, differentiation, and growth factor secretion potential. The functional decline is attributed to damaging extrinsic environmental factors and progressively worsening intrinsic genetic and biochemical processes. These ageing-associated deteriorative changes have been extensively documented, paving the way for the discovery of novel biomarkers of ageing for detection, diagnosis, and treatment of age-related diseases. Age-dependent changes in adult stem cells include numerical decline, loss of heterogeneity, reduced self-renewal and differentiation, leading to a drastic reduction in regenerative potential, and thereby drive the ageing process. Conversely, ageing also adversely alters the stem cell niche, disrupting the molecular pathways underlying stem cell homing, self-renewal, differentiation, and growth factor secretion, all of which are critical for tissue repair and regeneration. A holistic understanding of these molecular mechanisms, through empirical research and clinical trials, is essential for designing targeted therapies to modulate ageing and improve health parameters in older individuals.
Collapse
Affiliation(s)
- Jitendra Kumar Chaudhary
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi-110067, India; Department of Zoology, Shivaji College, University of Delhi, New Delhi-110027, India.
| | - Ajay Kumar Danga
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi-110067, India; National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi - 110067, India.
| | - Anita Kumari
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi-110067, India.
| | - Akshay Bhardwaj
- Global Research Alliances, Ashoka University, Rajiv Gandhi Education City, Sonepat, Haryana 131029, India.
| | - Pramod C Rath
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi-110067, India.
| |
Collapse
|
4
|
Ishihara T, Tsugawa H, Iwanami S, Chang JC, Minoda A, Arita M. Transcriptomic and lipidomic analysis of aging-associated inflammatory signature in mouse liver. Inflamm Regen 2025; 45:13. [PMID: 40319315 PMCID: PMC12049063 DOI: 10.1186/s41232-025-00377-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/20/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Aging-associated dysbiosis leads to chronic inflammation and the development of a range of aging-related diseases. The gut microbiota crosstalks with the host by providing lipid metabolites and modulating metabolic functions. However, the precise mechanism by which the gut microbiota regulates aging is unknown. The objective of this study was to examine the impact of the gut microbiota on the transcriptome and lipidome associated with aging in mouse liver. METHODS RNA-sequencing was conducted on the livers of young and aged male and female-specific pathogen-free (SPF) and germ-free (GF) mice to comprehensively analyze transcriptomic alterations with aging. We also reanalyzed our previously reported results on aging-associated changes in the hepatic lipidome to investigate the gut microbiota-dependent hepatic lipidome signatures associated with aging. RESULTS In contrast to the findings in male mice, the changes in hepatic transcriptome associated with aging were attenuated in female GF mice compared with those in SPF mice. In particular, the gene sets associated with inflammatory signatures (i.e., inflammation and tissue remodeling) were found to be suppressed in female GF mice. The ChIP-Atlas database predicted that transcription factors associated with sex differences may be involved in the gene signature of aged female GF mice. Significant differences in the lipid profile were observed between aged SPF and GF female mice, including in bile acids, sterol sulfates, lysophospholipids, oxidized triacylglycerols, vitamin D, and phytoceramides. Moreover, notable alterations were identified in the quality of phospholipids and sphingolipids. Integrated transcriptomic and lipidomic analysis identified candidate enzymes responsible for the change of lipid profiles in aged female mice. CONCLUSIONS The findings of this study offer new insights into the molecular mechanisms through which the gut microbiota regulates aging-related phenotypes such as inflammation in the liver, possibly through modulating lipid metabolism in a sex-dependent manner.
Collapse
Affiliation(s)
- Tomoaki Ishihara
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan.
- Faculty of Pharmaceutical Sciences, Nagasaki International University, 2825-7, Huis Ten Bosch, Sasebo, Nagasaki, 859-3298, Japan.
| | - Hiroshi Tsugawa
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei-shi, Tokyo, 184-8588, Japan
- Metabolome Informatics Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- Molecular and Cellular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Seigo Iwanami
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Jen-Chien Chang
- Laboratory for Cellular Epigenomics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Aki Minoda
- Laboratory for Cellular Epigenomics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
- Department of Human Biology, Radboud Institute for Molecular Life Sciences, Faculty of Science, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan.
- Molecular and Cellular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan.
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan.
| |
Collapse
|
5
|
Xue W, Guo N, Shan L, Zhang Z, Sun Y, Wang Y, Fang X, Liu X, Liu J, Hu C. Caveolin-1 protects against liver injury and lipid accumulation in alcoholic fatty liver via ferroptosis resistance. Mol Immunol 2025; 181:53-65. [PMID: 40073697 DOI: 10.1016/j.molimm.2025.02.012] [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: 03/03/2024] [Revised: 12/12/2024] [Accepted: 02/19/2025] [Indexed: 03/14/2025]
Abstract
Alcoholic fatty liver (AFL) is one of the most common chronic liver diseases globally with complex and controversial pathogenesis. Recent evidence suggests that iron overload and lipid peroxidation are risk factors for AFL. Caveolin-1 (CAV1) is an important signal platform that can maintain lipid homeostasis during the development of non-alcoholic fatty liver. Here, we studied the effect of CAV1 on ferroptosis in AFL. The AFL mouse model was established by chronic-plus-binge alcohol feeding. In vitro, AML-12 cells were incubated with ethanol and oleic acid for 48 h. We found alcohol-induced AFL triggered ferroptosis and decreased CAV1 expression. Overexpression of CAV1 by CAV1 scaffolding domain peptides (CSD) attenuated liver injury and hepatic steatosis, as well as inhibited ferroptosis in AFL mice. Additionally, the effects of CAV1 on ferroptosis-related protein levels (such as SLC7A11, GPX4, and ACSL4) and lipid accumulation were reversed by its small interfering RNA administration. Ferroptosis agonist (Erastin) treatment abrogated CAV1 plasmid-mediated ferroptosis resistance and steatosis alleviation. Collectively, the results revealed a crucial role of CAV1 in preventing hepatic steatosis and ferroptosis in alcohol-induced liver injury, which may identify potential targets for the treatment of AFL.
Collapse
Affiliation(s)
- Weiju Xue
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Ning Guo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Liang Shan
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Zhengsheng Zhang
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Yuquan Sun
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Yong Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Xing Fang
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Xiuzhen Liu
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Jianjun Liu
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China.
| | - Chengmu Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
6
|
Zhang Z, Luo Y, Zhang H, Zeng Z, Zheng W, Zhao Y, Huang Y, Shen L. Exploring the mechanisms of cow placental peptides in delaying liver aging based on mitochondrial energy metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119593. [PMID: 40064320 DOI: 10.1016/j.jep.2025.119593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Placenta is a kind of traditional Chinese medicine, known as "Ziheche". The role of cow placental peptides (CPP) in delaying liver aging has been reported, and in-depth exploration of the specific regulatory mechanisms is of great significance for the recycling and utilization of CPP and the development of natural anti-aging drugs. AIM OF THE STUDY To investigate the protective effects and mechanisms of CPP on liver aging induced by D-galactose (D-gal) in mice from the perspective of mitochondrial energy metabolism. METHODS An aging model was induced in mice using D-gal. The body weight and liver index of mice were measured, followed by staining and electron microscopy to observe liver morphology and aging markers. Reactive oxygen species (ROS) levels and antioxidant-related indicators were assessed, and mitochondrial function was evaluated. Finally, changes and mechanisms in liver transcriptomics and targeted mitochondrial energy metabolomics were analyzed and integrated to elucidate the regulatory pathways through which CPP delays liver aging. RESULTS CPP improved liver structural damage, oxidative stress, and mitochondrial dysfunction induced by D-galactose in aging mice. It increased the final body weight and liver index, alleviated hepatocyte swelling and degeneration, enhanced liver antioxidant capacity, and restored normal mitochondrial morphology and function. The combined analysis of targeted mitochondrial energy metabolomics and liver transcriptomics revealed that CPP directly or indirectly regulated mitochondrial energy metabolism and delayed aging by influencing the cAMP signaling pathway, PI3K-Akt signaling pathway, oxidative phosphorylation, and other pathways, thereby modulating related genes and metabolites.
Collapse
Affiliation(s)
- Zeru Zhang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yuxin Luo
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Hanwen Zhang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhi Zeng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Weijian Zheng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yuquan Zhao
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yixin Huang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Liuhong Shen
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
7
|
Lee DY, Noren Hooten N, O'Connell JF, Lee BY, Kim Y. The Role of Ginseng and Its Bioactive Compounds in Aging: Cells and Animal Studies. Annu Rev Food Sci Technol 2025; 16:333-354. [PMID: 39971378 DOI: 10.1146/annurev-food-111523-121753] [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: 02/21/2025]
Abstract
Aging is an inevitable process that is characterized by physiological deterioration and increased vulnerability to stressors. Therefore, the interest in hallmarks, mechanisms, and ways to delay or prevent aging has grown for decades. Natural plant products and their bioactive compounds have been studied as a promising strategy to overcome aging. Ginseng, a traditional herbal medicine, and its bioactive compound, the ginsenosides, have increasingly gained attention because of various pharmacological functions. This review introduces the species, useful parts, characteristics, and active components of ginseng. It primarily focuses on the bioconversion of ginsenosides through the unique steaming and drying process. More importantly, this review enumerates the antiaging mechanisms of ginseng, ginsenosides, and other bioactive compounds, highlighting their potential to extend the health span and mitigate age-related diseases based on twelve representative hallmarks of aging.
Collapse
Affiliation(s)
- Da-Yeon Lee
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, Oklahoma, USA;
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, Maryland, USA
| | - Jennifer F O'Connell
- Center for Scientific Review, National Institutes of Health, Bethesda, Maryland, USA
| | - Boo-Yong Lee
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea;
| | - Yoo Kim
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, Oklahoma, USA;
| |
Collapse
|
8
|
Langhans B, Strassburg CP, Röcken C, Kalthoff S. A Common UDP-Glucuronosyltransferase ( UGT) 1A Haplotype Is Associated With Accelerated Aging in Humanized Transgenic Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2025; 2025:3203439. [PMID: 40182762 PMCID: PMC11968170 DOI: 10.1155/omcl/3203439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025]
Abstract
Background: Aging is characterized by the progressive decline of physiological functions and is associated with an increasing risk for developing multiple age-related diseases. UDP-glucuronosyltransferase (UGT)1A enzymes detoxify a variety of endo- and xenobiotic reactive metabolites, thereby acting as indirect antioxidants. A common genetic UGT1A haplotype was shown to affect redox balance in humanized transgenic (htg) UGT1A mice. Since oxidative stress is a main activator of cellular senescence, we aimed to investigate the role of genetic UGT1A variants in the process of aging. Methods: HtgUGT1A-WT and htgUGT1A-SNP mice were harvested at the age of either 12 weeks (young) or 18 months (aged). The effect of aging was examined by analyzing UGT1A expression and activity, expression of senescence markers, and senescence-associated secretory phenotype (SASP) factors, as well as blood counts, serum parameter, and histological staining. Results: In comparison to aged htgUGT1A-WT mice, hepatic UGT1A mRNA and protein expression as well as UGT activity were significantly reduced in aged htgUGT1A-SNP mice. Moreover, elderly htgUGT1A-SNP mice exhibited increased levels of oxidative stress, senescence markers, SASP factors, and peripheral leukocyte counts compared to the respective htgUGT1A-WT mice. Consistent with these findings, we observed higher amounts of collagen and amyloid fibrils as well as an elevated senescence-associated β-galactosidase (SA-β-gal) activity in histological sections of the liver obtained from aged htgUGT1A-SNP mice. Conclusion: Our data suggest an accelerated aging process caused by a common UGT1A haplotype. Moreover, elderly individuals carrying the UGT1A haplotype might exhibit an altered metabolism of drugs, which could necessitate dose adjustments.
Collapse
Affiliation(s)
- Bettina Langhans
- Department of Internal Medicine I, University Hospital Bonn 53127, Bonn, Germany
| | | | - Christoph Röcken
- Department of Pathology, University Hospital Schleswig-Holstein 24105, Kiel, Germany
| | - Sandra Kalthoff
- Department of Internal Medicine I, University Hospital Bonn 53127, Bonn, Germany
| |
Collapse
|
9
|
Mohallem R, Schaser AJ, Aryal UK. Proteomic and phosphoproteomic signatures of aging mouse liver. GeroScience 2025:10.1007/s11357-025-01601-0. [PMID: 40087212 DOI: 10.1007/s11357-025-01601-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/04/2025] [Indexed: 03/17/2025] Open
Abstract
The liver is a metabolic powerhouse, crucial for regulating carbohydrates, fats, and protein metabolism. In this study, we conducted a comparative proteomic and phosphoproteomic analysis of aging mouse livers from young adults (3-4 months) and old (19-21 months) mice to identify age-related changes in liver proteins and phosphosites, which were linked to various metabolic pathways. In old mice, proteins associated with the "complement and coagulation cascade," "age-rage signaling in diabetic complications," and "biosynthesis of unsaturated fatty acids" were increased, while those linked to "oxidative phosphorylation," "steroid hormone biosynthesis," and "tryptophan metabolism" were decreased. Interestingly, aging was marked by a significant decrease in liver protein phosphorylation, with nearly 90% of significant phosphosites being downregulated. Pathway analysis of the downregulated phosphosites highlighted connections to "non-small cell lung cancer," "lysine degradation," "cell differentiation," and "glycerophospholipid metabolism." Decreased phosphorylation of several kinases that are linked to cell proliferation, particularly those in the MAPK signaling pathway, including Erk1, EGFR, RAF1, and BRAF was also observed highlighting their important role in the liver. This study identified an important relationship between proteins, phosphosites, and their connections to known as well as new pathways, expanding upon our current knowledge and providing a basis for future studies focused on age-related metabolic traits.
Collapse
Affiliation(s)
- Rodrigo Mohallem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Allison J Schaser
- Department of Speech, Language, and Hearing Sciences, Purdue University, West Lafayette, IN, 47907, USA.
| | - Uma K Aryal
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA.
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
10
|
Ngcobo NN. Influence of Ageing on the Pharmacodynamics and Pharmacokinetics of Chronically Administered Medicines in Geriatric Patients: A Review. Clin Pharmacokinet 2025; 64:335-367. [PMID: 39798015 PMCID: PMC11954733 DOI: 10.1007/s40262-024-01466-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2024] [Indexed: 01/13/2025]
Abstract
As people age, the efficiency of various regulatory processes that ensure proper communication between cells and organs tends to decline. This deterioration can lead to difficulties in maintaining homeostasis during physiological stress. This includes but is not limited to cognitive impairments, functional difficulties, and issues related to caregivers which contribute significantly to medication errors and non-adherence. These factors can lead to higher morbidity, extended hospital stays, reduced quality of life, and even mortality. The decrease in homeostatic capacity varies among individuals, contributing to the greater variability observed in geriatric populations. Significant pharmacokinetic and pharmacodynamic alterations accompany ageing. Pharmacokinetic changes include decreased renal and hepatic clearance and an increased volume of distribution for lipid-soluble drugs, which prolong their elimination half-life. Pharmacodynamic changes typically involve increased sensitivity to various drug classes, such as anticoagulants, antidiabetic and psychotropic medications. This review examines the primary age-related physiological changes in geriatrics and their impact on the pharmacokinetics and pharmacodynamics of medications.
Collapse
Affiliation(s)
- Nokwanda N Ngcobo
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
11
|
Zhang M, Wei J, Sun Y, He C, Ma S, Pan X, Zhu X. The efferocytosis process in aging: Supporting evidence, mechanisms, and therapeutic prospects for age-related diseases. J Adv Res 2025; 69:31-49. [PMID: 38499245 PMCID: PMC11954809 DOI: 10.1016/j.jare.2024.03.008] [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: 12/16/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Aging is characterized by an ongoing struggle between the buildup of damage caused by a combination of external and internal factors. Aging has different effects on phagocytes, including impaired efferocytosis. A deficiency in efferocytosis can cause chronic inflammation, aging, and several other clinical disorders. AIM OF REVIEW Our review underscores the possible feasibility and extensive scope of employing dual targets in various age-related diseases to reduce the occurrence and progression of age-related diseases, ultimately fostering healthy aging and increasing lifespan. Key scientific concepts of review Hence, the concurrent implementation of strategies aimed at augmenting efferocytic mechanisms and anti-aging treatments has the potential to serve as a potent intervention for extending the duration of a healthy lifespan. In this review, we comprehensively discuss the concept and physiological effects of efferocytosis. Subsequently, we investigated the association between efferocytosis and the hallmarks of aging. Finally, we discuss growing evidence regarding therapeutic interventions for age-related disorders, focusing on the physiological processes of aging and efferocytosis.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yu Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Chang He
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shiyin Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
12
|
Zhang Y, Gong C, Tao L, Zhai J, Huang F, Zhang S. Involvement of SIRT1-mediated aging in liver diseases. Front Cell Dev Biol 2025; 13:1548015. [PMID: 40052151 PMCID: PMC11882576 DOI: 10.3389/fcell.2025.1548015] [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: 12/19/2024] [Accepted: 01/27/2025] [Indexed: 03/09/2025] Open
Abstract
Liver disease is a significant global health issue, responsible for millions of deaths annually. Aging, characterized by the gradual decline in cellular and physiological functions, impairs tissue regeneration, increases susceptibility to liver diseases, and leads to a decline in liver health. Silent information regulator 1 (SIRT1), a NAD⁺-dependent deacetylase, has emerged as a pivotal factor in modulating age-related changes in the liver. SIRT1 preserves liver function by regulating essential aging-related pathways, including telomere maintenance, epigenetic modifications, cellular senescence, intercellular communication, inflammation, and mitochondrial function. Notably, SIRT1 levels naturally decline with age, contributing to liver disease progression and increased vulnerability to injury. This review summarizes the regulatory role of SIRT1 in aging and its impact on liver diseases such as liver fibrosis, alcoholic associated liver disease (ALD), metabolic dysfunction-associated steatotic liver disease (MASLD), and metabolic dysfunction-associated steatohepatitis (MASH), hepatocellular carcinoma (HCC). We also discuss emerging therapeutic approaches, including SIRT1 activators, gene therapy, and nutritional interventions, which are evaluated for their potential to restore SIRT1 function and mitigate liver disease progression. Finally, we highlight future research directions to optimize SIRT1-targeted therapies for clinical applications in age-related liver conditions.
Collapse
Affiliation(s)
- Yueming Zhang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, China
| | - Chang Gong
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, China
| | - Lina Tao
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, China
| | - Jinghui Zhai
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, China
| | - Fengwei Huang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, China
- College of Pharmacy, Jilin University, Changchun, Jilin, China
| | - Sixi Zhang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, China
- College of Pharmacy, Jilin University, Changchun, Jilin, China
| |
Collapse
|
13
|
Mo H, Yue P, Li Q, Tan Y, Yan X, Liu X, Xu Y, Luo Y, Palihati S, Yi C, Zhang H, Yuan M, Yang B. The role of liver sinusoidal endothelial cells in metabolic dysfunction-associated steatotic liver diseases and liver cancer: mechanisms and potential therapies. Angiogenesis 2025; 28:14. [PMID: 39899173 DOI: 10.1007/s10456-025-09969-5] [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: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 02/04/2025]
Abstract
Liver sinusoidal endothelial cells (LSECs), with their unique morphology and function, have garnered increasing attention in chronic liver disease research. This review summarizes the critical roles of LSECs under physiological conditions and in two representative chronic liver diseases: metabolic dysfunction-associated steatotic liver disease (MASLD) and liver cancer. Under physiological conditions, LSECs act as selective barriers, regulating substance exchange and hepatic blood flow. Interestingly, LSECs exhibit contrasting roles at different stages of disease progression: in the early stages, they actively resist disease advancement and help restore sinusoidal homeostasis; whereas in later stages, they contribute to disease worsening. During this transition, LSECs undergo capillarization, lose their characteristic markers, and become dysfunctional. As the disease progresses, LSECs closely interact with hepatocytes, hepatic stellate cells, various immune cells, and tumor cells, driving processes such as steatosis, inflammation, fibrosis, angiogenesis, and carcinogenesis. Consequently, targeting LSECs represents a promising therapeutic strategy for chronic liver diseases. Relevant therapeutic targets and potential drugs are summarized in this review.
Collapse
Affiliation(s)
- Hanjun Mo
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Pengfei Yue
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Qiaoqi Li
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Yinxi Tan
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
| | - Xinran Yan
- Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyue Liu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuanwei Xu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yingzhe Luo
- Department of Medical Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Chengdu, 610075, Sichuan, China
| | - Suruiya Palihati
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Cheng Yi
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Hua Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, No. 20, Section 3, South Renmin Road, Chengdu, 610041, China.
- Key Laboratory of Chronobiology (Sichuan University), National Health Commission of China, Chengdu, 610041, China.
| | - Minlan Yuan
- Mental Health Center and Psychiatric Laboratory, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China.
| | - Biao Yang
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
14
|
Xie P, Ommati MM, Chen D, Chen W, Han L, Zhao X, Wang H, Xu S, Sun P. Hepatotoxic effects of environmentally relevant concentrations of polystyrene microplastics on senescent Zebrafish (Danio rerio): Patterns of stress response and metabolomic alterations. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 279:107252. [PMID: 39847840 DOI: 10.1016/j.aquatox.2025.107252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/07/2025] [Accepted: 01/15/2025] [Indexed: 01/25/2025]
Abstract
The hepatotoxicity of microplastics (MPs) has garnered increasing attention, but their effects on elderly organisms remain inadequately characterized, particularly concerning hepatic stress response patterns in environmental conditions. In this study, a 10-day exposure period of elderly zebrafish to polystyrene microplastics (PS-MPs, 1 µm) was conducted, with exposure concentrations set at 5.6 × 10-7 µg/L, 5.6 × 10-4 µg/L, and 5.6 × 10-1 µg/L. PS-MPs-induced toxicity varied with concentration: superoxide dismutase (SOD), complement 3 (C3), and complement 4 (C4) initially decreased before rising; 8‑hydroxy-2-deoxyguanosine (8-OhdG), interleukin-6 (IL-6), and interleukin-8 (IL-8) increased at high concentrations. Additionally, catalase (CAT) activity and thiobarbituric acid reactive substances (TBARS) contents rose with concentration. The aged zebrafish liver exhibited differentiation driven by responsiveness; low levels cause homeostatic disruption, and high levels induce genotoxicity and immune activation. LC-MS identified twelve crucial metabolites involved in 18 metabolic pathways, including amino acids (L-tyrosine, l-arginine), lipids (phospholipids, 12(S)-leukotriene B4 and triglycerides), and N-acetylneuraminic acid, related to energy, immunity, and neurological health. Overall, elderly zebrafish exhibited clear dose-dependent thresholds and distinct physiological stress responses under varying concentrations of PS-MPs. These findings reveal how PS-MP exposure can affect physiological health and metabolism, offering critical insights into the ecological risks faced by aging organisms.
Collapse
Affiliation(s)
- Pengfei Xie
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Mohammad Mehdi Ommati
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Deshan Chen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Weijun Chen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Lei Han
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Xinquan Zhao
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai 810016, China
| | - Hongwei Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China
| | - Shixiao Xu
- Northwest Institute of Plateau Biology Chinese Academy of Sciences, Xining, Qinghai 810008, China
| | - Ping Sun
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471003, China.
| |
Collapse
|
15
|
Zapatería B, Arias E. Aging, cancer, and autophagy: connections and therapeutic perspectives. Front Mol Biosci 2025; 11:1516789. [PMID: 39935707 PMCID: PMC11811537 DOI: 10.3389/fmolb.2024.1516789] [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: 10/24/2024] [Accepted: 12/24/2024] [Indexed: 02/13/2025] Open
Abstract
Aging and cancer are intricately linked through shared molecular processes that influence both the onset of malignancy and the progression of age-related decline. As organisms age, cellular stress, genomic instability, and an accumulation of senescent cells create a pro-inflammatory environment conducive to cancer development. Autophagy, a cellular process responsible for degrading and recycling damaged components, plays a pivotal role in this relationship. While autophagy acts as a tumor-suppressive mechanism by preventing the accumulation of damaged organelles and proteins, cancer cells often exploit it to survive under conditions of metabolic stress and treatment resistance. The interplay between aging, cancer, and autophagy reveals key insights into tumorigenesis, cellular senescence, and proteostasis dysfunction. This review explores the molecular connections between these processes, emphasizing the potential for autophagy-targeted therapies as strategies that could be further explored in both aging and cancer treatment. Understanding the dual roles of autophagy in suppressing and promoting cancer offers promising avenues for therapeutic interventions aimed at improving outcomes for elderly cancer patients while addressing age-related deterioration.
Collapse
Affiliation(s)
- Begoña Zapatería
- Department of Medicine (Marion Bessin Liver Research Center), Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Esperanza Arias
- Department of Medicine (Marion Bessin Liver Research Center), Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States
- Einstein Aging Research Center, Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
16
|
Li H, Zhou Y, Cai C, Liang H, Li X, Huang M, Fan S, Bi H. Fenofibrate induces liver enlargement in aging mice via activating the PPARα-YAP signaling pathway. Chem Biol Interact 2025; 405:111286. [PMID: 39442682 DOI: 10.1016/j.cbi.2024.111286] [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: 09/06/2024] [Revised: 10/12/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024]
Abstract
Fenofibrate is a clinically prescribed drug for treating hypertriglyceridemia, which is also a classic peroxisome proliferator-activated receptor α (PPARα) agonist. We previously reported that fenofibrate induced liver enlargement in adult mice partially through activation of the yes-associated protein (YAP) signaling pathway. However, the effects of fenofibrate on liver enlargement and the YAP signaling pathway in aging mice remain unclear. In this study, D-galactose-induced aging mice, naturally aging mice, and senescence-accelerated mice P8 (SAMP8) were used to investigate the effects of aging on fenofibrate-induced liver enlargement and YAP signaling activation. The results showed that fenofibrate-induced liver enlargement in aging mice was consistent with that of adult mice. The effects of fenofibrate on hepatocyte enlargement around the central vein (CV) area and hepatocyte proliferation around the portal vein (PV) area were comparable between adult and aging mice. There was no significant difference in the upregulation of PPARα downstream proteins between the two groups following fenofibrate treatment. Fenofibrate treatment also increased the expression of proliferation-related proteins and activated the YAP signaling pathway to a similar degree in both groups. In summary, these results demonstrate that the fenofibrate-induced liver enlargement and activation of the YAP pathway are consistent between adult and aging mice, indicating that the effects of fenofibrate on promoting liver enlargement and its activation of the PPARα and YAP pathway were independent of aging. These findings offer a new perspective for the clinical use of fenofibrate in elderly patients and provide a new insight for the role of PPARα in liver enlargement.
Collapse
Affiliation(s)
- Huilin Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Yanying Zhou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Chenghui Cai
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Hangfei Liang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Xuan Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Shicheng Fan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| | - Huichang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China; The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, 518055, China.
| |
Collapse
|
17
|
De Matteis C, Crudele L, Di Buduo E, Cantatore S, Gadaleta RM, Cariello M, Suppressa P, Antonica G, Berardi E, Graziano G, Moschetta A. Hyperhomocysteinemia is linked to MASLD. Eur J Intern Med 2025; 131:49-57. [PMID: 39482164 DOI: 10.1016/j.ejim.2024.10.014] [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] [Received: 05/15/2024] [Revised: 09/14/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024]
Abstract
BACKGROUND AND AIMS Homocysteine (Hcy) levels are elevated in different conditions, including cardiovascular diseases (CVD), diabetes, and metabolic-associated steatotic liver disease (MASLD). In this observational retrospective study, we analyzed Hcy levels in a population of 901 outpatients, considering its putative etiological role in MASLD. METHODS A total of 901 outpatients underwent physical and biochemical evaluations. Abdominal and carotid ultrasound were performed to assess liver steatosis, carotid intima-media thickness (IMT) and presence of atherosclerotic plaque. RESULTS Hyperhomocysteinemia (HHcy) was identified in 140 subjects (16 %). Patients with HHcy showed glucose metabolism impairment (p < 0.001), altered lipid profile (p < 0.001), low Vitamin D levels (p < 0.0001), increased cardiovascular risk (p < 0.001). We then investigated the relationship between Hcy and MASLD (OR=3.6, p < 0.0001), finding that the relationship remained significant also when accounting for confounding variables (age, sex) (OR=3.2, p < 0.0001). Hcy values were significantly higher (p < 0.0001) in patients with MASLD (n = 78, 29.4 ± 10.1μmol/l) compared to those without MASLD (20.4 ± 4.8 1μmol/l). Furthermore, in MASLD patients we found a direct correlation between Hcy level and waist circumference (r = 0.3, p < 0.001) and an inverse correlation with both HDL-c (r=-0.4, p < 0.001) and Vitamin D levels (r=-0.24, p < 0.05). CONCLUSIONS Our data suggest an intriguing scenario whereby HHcy is present in patients with MASLD and is associated to lower vitamin D and altered glucose and lipid profile. Thus, considering Hcy levels may help clinicians with the management of patients with increased MASLD risk.
Collapse
Affiliation(s)
- Carlo De Matteis
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Lucilla Crudele
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Ersilia Di Buduo
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Salvatore Cantatore
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | | | - Marica Cariello
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Patrizia Suppressa
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Gianfranco Antonica
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Elsa Berardi
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Giusi Graziano
- Center for Outcomes Research and Clinical Epidemiology (CORESEARCH), 65124 Pescara, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy; INBB National Institute for Biostructure and Biosystems, Viale delle Medaglie d'Oro 305, 00136 Roma, Italy.
| |
Collapse
|
18
|
Zhang C, Fu Z, Zhang R. Reduced expressions of TCA cycle genes during aging in humans and mice. Biochem Biophys Res Commun 2024; 738:150917. [PMID: 39504716 PMCID: PMC11583999 DOI: 10.1016/j.bbrc.2024.150917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024]
Abstract
Aging is associated with a decline in physiological functions and an increased risk of metabolic disorders. The liver, a key organ in metabolism, undergoes significant changes during aging that can contribute to systemic metabolic dysfunction. This study investigates the expression of genes involved in the tricarboxylic acid (TCA) cycle, a critical pathway for energy production, in the aging liver. We analyzed RNA sequencing data from the Genotype-Tissue Expression (GTEx) project to assess age-related changes in gene expression in the human liver. To validate our findings, we conducted complementary studies in young and old mice, examining the expression of key TCA cycle genes using quantitative real-time PCR. Our analysis of the GTEx dataset revealed a significant reduction in the expression of many genes that are critical for metabolism, including fat mass and obesity associated (FTO) and adiponectin receptor 1 (ADIPOR1). The most overrepresented pathway among the statistically enriched ones was the TCA cycle, with multiple genes exhibiting downregulation in older humans. This reduction was consistent with findings in aging mice, which also showed decreased expression of several TCA cycle genes. These results suggest a conserved pattern of age-related downregulation of TCA cycle, potentially leading to diminished mitochondrial function and energy production in the liver. The reduced expression of TCA cycle genes in the aging liver may contribute to metabolic dysfunction and increased susceptibility to age-related diseases. Understanding the molecular basis of these changes provides new insights into the aging process and highlights potential targets for interventions aimed at promoting healthy aging and preventing metabolic disorders.
Collapse
Affiliation(s)
- Chao Zhang
- Biostatistics Shared Resource, Winship Cancer Institute of Emory University, 718 Gatewood Rd. NE, Atlanta, GA, 30322, USA
| | - Zhiyao Fu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Ren Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| |
Collapse
|
19
|
Rahimi Naiini M, Shahouzehi B, Khaksari M, Azizi S, Naghibi N, Nazari-Robati M. Ellagic acid reduces hepatic lipid contents through regulation of SIRT1 and AMPK in old rats. Arch Physiol Biochem 2024; 130:686-693. [PMID: 37814948 DOI: 10.1080/13813455.2023.2262165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/07/2023] [Indexed: 10/11/2023]
Abstract
OBJECTIVE Ellagic acid is used in traditional medicine for the treatment of lipid disorders. In this study, the effects of ellagic acid on key regulators of lipid metabolism, and histopathological alterations in aged liver were examined. METHODS A total of 21 male Wistar rats were divided into three groups, including young control, old control, and old ellagic acid. After one month of treatment with ellagic acid, the expression levels of hepatic SIRT1, AMPK, SREBP-1c, PPAR-α, and phosphorylated AMPK (p-AMPK) were evaluated. The levels of several serum biochemical factors, and hepatic triglyceride, and cholesterol contents were assessed. RESULTS Ellagic acid elevated the levels of SIRT1, p-AMPK, and PPAR-α and reduced SREBP-1c level in the liver of old rats. It decreased triglyceride and cholesterol contents in the aged liver and improved histopathological changes. CONCLUSIONS The results demonstrated that ellagic acid can exert protective effects against hepatic lipid metabolism disorders induced by ageing.
Collapse
Affiliation(s)
- Mahdis Rahimi Naiini
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Beydolah Shahouzehi
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahrzad Azizi
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Niloufar Naghibi
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdieh Nazari-Robati
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
20
|
Tello-Palencia MA, Yang T, Sularz O, Demers LE, Ma Y, Boycott C, Zhang HA, Lubecka-Gajewska K, Kumar S, Ramsey BS, Torregrosa-Allen S, Elzey BD, Lanman NA, Korthauer K, Stefanska B. Pterostilbene Targets Hallmarks of Aging in the Gene Expression Landscape in Blood of Healthy Rats. Mol Nutr Food Res 2024; 68:e2400662. [PMID: 39562169 DOI: 10.1002/mnfr.202400662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/05/2024] [Indexed: 11/21/2024]
Abstract
SCOPE Polyphenols from the phytoestrogen group, including pterostilbene (PTS), are known for their antioxidant, anti-inflammatory, and anti-cancer effects. In recent reports, phytoestrogens attenuate age-related diseases; however, their pro-longevity effects in healthy models in mammals remain unknown. As longevity research demonstrates age-related transcriptomic signatures in human blood, the current study hypothesizes that phytoestrogen-supplemented diet may induce changes in gene expression that ultimately confer pro-longevity benefits. METHODS AND RESULTS In the present study, RNA sequencing is conducted to determine transcriptome-wide changes in gene expression in whole blood of healthy rats consuming diets supplemented with phytoestrogens. Ortholog cell deconvolution is applied to analyze the omics data. The study discovered that PTS leads to changes in the gene expression landscape and PTS-target genes are associated with functions counteracting hallmarks of aging, including genomic instability, epigenetic alterations, compromised autophagy, mitochondrial dysfunction, deregulated nutrient sensing, altered intercellular interaction, and loss of proteostasis. These functions bridge together under anti-inflammatory effects through multiple pathways, including immunometabolism, where changes in cellular metabolism (e.g., ribosome biogenesis) impact the immune system. CONCLUSION The findings provide a rationale for pre-clinical and clinical longevity studies and encourage investigations on PTS in maintaining cellular homeostasis, decelerating the process of aging, and improving conditions with chronic inflammation.
Collapse
Affiliation(s)
- Marco A Tello-Palencia
- Department of Statistics, Faculty of Science, the University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Tony Yang
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Olga Sularz
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
- Department of Human Nutrition and Dietetics, Faculty of Food Technology, University of Agriculture in Krakow, Krakow, 31-120, Poland
| | - Louis Erik Demers
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
- Department of Parasitology, McGill University, Montreal, QC, H9X 3V9, Canada
| | - Yuexi Ma
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Cayla Boycott
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Huiying Amelie Zhang
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | | | - Sadhri Kumar
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Benjamin S Ramsey
- Purdue University Institute for Cancer Research, Purdue University, West Lafayette, IN, 47906, USA
| | - Sandra Torregrosa-Allen
- Purdue University Institute for Cancer Research, Purdue University, West Lafayette, IN, 47906, USA
| | - Bennett D Elzey
- Purdue University Institute for Cancer Research, Purdue University, West Lafayette, IN, 47906, USA
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Nadia Atallah Lanman
- Purdue University Institute for Cancer Research, Purdue University, West Lafayette, IN, 47906, USA
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Keegan Korthauer
- Department of Statistics, Faculty of Science, the University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Vancouver, BC, V6H 0B3, Canada
| | - Barbara Stefanska
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, the University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| |
Collapse
|
21
|
Pinto AP, Muñoz VR, Tavares MEA, Neto IVDS, Dos Santos JR, Rodrigues GS, Carolino ROG, Alberici LC, Simabuco FM, Teixeira GR, Pauli JR, de Moura LP, Cintra DE, Ropelle ER, Freitas EC, Rivas DA, da Silva ASR. Short-term exercise counteracts accelerated ageing impacts on physical performance and liver health in mice. Clin Exp Pharmacol Physiol 2024; 51:e70001. [PMID: 39477552 DOI: 10.1111/1440-1681.70001] [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: 07/02/2024] [Revised: 09/15/2024] [Accepted: 10/01/2024] [Indexed: 01/06/2025]
Abstract
Senescence impairs liver physiology, mitochondrial function and circadian regulation, resulting in systemic metabolic dysregulation. Given the limited research on the effects of combined exercise on an ageing liver, this study aimed to evaluate its impact on liver metabolism, circadian rhythms and mitochondrial function in senescence-accelerated mouse-prone 8 (SAMP8) and senescence-accelerated mouse-resistant 1 (SAMR1) mice. Histological, reverse transcription quantitative polymerase chain reaction (RT-qPCR) and immunoblotting analyses were conducted, supplemented by transcriptomic data sets and AML12 hepatocyte studies. Sedentary SAMP8 mice exhibited decreased muscle strength, reduced mitochondrial complex I levels and increased lipid droplet accumulation. In contrast, combined exercise mitigated muscle strength loss, upregulated proteins involved in mitochondrial complexes (CIII, CIV, CV) and increased Bmal1 messenger RNA (mRNA) expression in the liver. These molecular adaptations are associated with healthier liver phenotypes and may influence metabolic function and cellular longevity. Notably, elevated lipid content in aged mice was reduced post-exercise, indicating liver benefits even after a relatively short intervention. The combined exercise regimen did not improve aerobic capacity, likely due to the low volume and brief duration of running. Moreover, no significant effects were observed in SAMR1 mice, possibly because the training intensity was insufficient for younger, healthier animals. These findings underscore the potential of combined strength and endurance exercise to attenuate age-related liver dysfunction, particularly in ageing populations.
Collapse
Affiliation(s)
- Ana P Pinto
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
| | - Vitor R Muñoz
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
| | - Maria Eduarda A Tavares
- Department of Physical Education, State University of São Paulo (UNESP), São Paulo, Brazil
- Multicentric Program of Postgraduate in Physiological Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil
| | - Ivo V de Sousa Neto
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
| | - Jonathas R Dos Santos
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo-FCFRP USP, São Paulo, Brazil
| | - Guilherme S Rodrigues
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| | - Ruither O Gomes Carolino
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
| | - Luciane C Alberici
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo-FCFRP USP, São Paulo, Brazil
| | - Fernando M Simabuco
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
- Department of Biochemistry, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Giovana R Teixeira
- Department of Physical Education, State University of São Paulo (UNESP), São Paulo, Brazil
- Multicentric Program of Postgraduate in Physiological Sciences, School of Dentistry of Araçatuba, São Paulo State University (UNESP), São Paulo, Brazil
| | - José R Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | - Leandro P de Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Dennys E Cintra
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | - Eduardo R Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), São Paulo, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, Brazil
| | - Ellen C Freitas
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| | - Donato A Rivas
- Center for Exercise Medicine Research, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, Virginia, USA
| | - Adelino S R da Silva
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), São Paulo, Brazil
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
22
|
Grigio V, Guerra LHA, Silva SB, Freitas MB, Taboga SR, Vilamaior PSL. Coconut oil affects aging-related changes in Mongolian gerbil liver morphophysiology. J Nutr Biochem 2024; 134:109749. [PMID: 39233189 DOI: 10.1016/j.jnutbio.2024.109749] [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: 03/26/2024] [Revised: 08/03/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
Aging causes changes in liver morphophysiology, altering hepatocyte morphology and organ function. Due to its antioxidant and anti-inflammatory properties, coconut oil has been used as a therapeutic agent in diets, in an attempt to attenuate alterations in the liver naturally caused by aging. Herein, we evaluated the effects of coconut oil consumption during aging on Mongolian gerbil liver morphophysiology. The animals were divided into three experimental groups: the gerbils in the Adult Control Group (AC) were euthanized at 3 months of age, the gerbils in the Old Control Group (OC) at 15 months of age, and the gerbils in the Coconut Oil Group (CO) received 0.1 ml/day of coconut oil for 12 months and were euthanized at 15 months of age. Prolonged consumption of coconut oil during aging prevented the animals and the liver from gaining mass. However, the other results showed that coconut oil intensified the morphophysiological alterations of aging, promoting an increase in the hepatocyte cytoplasm and nuclei. In addition, an increase in blood vessels, reticular fibers, lipid droplets, and lipofuscin granules were observed in the CO group. Finally, the results also demonstrated that coconut oil promotes an increase in lipid peroxidation, indicated by an increase in MDA levels. We therefore conclude that coconut oil has the potential to intensify the morphophysiological alterations that occur in the liver during aging.
Collapse
Affiliation(s)
- Vitor Grigio
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São Paulo, Brazil
| | - Luiz Henrique Alves Guerra
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São Paulo, Brazil
| | - Stella Bicalho Silva
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São Paulo, Brazil; Department of Animal Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | | | - Sebastião Roberto Taboga
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São Paulo, Brazil
| | - Patrícia Simone Leite Vilamaior
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São Paulo, Brazil.
| |
Collapse
|
23
|
Singh M, Ali H, Renuka Jyothi S, Kaur I, Kumar S, Sharma N, Siva Prasad GV, Pramanik A, Hassan Almalki W, Imran M. Tau proteins and senescent Cells: Targeting aging pathways in Alzheimer's disease. Brain Res 2024; 1844:149165. [PMID: 39155034 DOI: 10.1016/j.brainres.2024.149165] [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: 06/16/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease characterized by abnormal accumulation of tau proteins and amyloid-β, leading to neuronal death and cognitive impairment. Recent studies have implicated aging pathways, including dysregulation of tau and cellular senescence in AD pathogenesis. In AD brains, tau protein, which normally stabilizes microtubules, becomes hyperphosphorylated and forms insoluble neurofibrillary tangles. These tau aggregates impair neuronal function and are propagated across the brain's neurocircuitry. Meanwhile, the number of senescent cells accumulating in the aging brain is rising, releasing a pro-inflammatory SASP responsible for neuroinflammation and neurodegeneration. This review explores potential therapeutic interventions for AD targeting tau protein and senescent cells, and tau -directed compounds, senolytics, eliminating senescent cells, and agents that modulate the SASP-senomodulators. Ultimately, a combined approach that incorporates tau-directed medications and targeted senescent cell-based therapies holds promise for reducing the harmful impact of AD's shared aging pathways.
Collapse
Affiliation(s)
- Mahaveer Singh
- School of Pharmacy and Technology Management, SVKMs NMIMS University, Shirpur campus, Maharastra India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan-303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali 140307, Punjab, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
24
|
Chanda F, Lin KX, Chaurembo AI, Huang JY, Zhang HJ, Deng WH, Xu YJ, Li Y, Fu LD, Cui HD, Shu C, Chen Y, Xing N, Lin HB. PM 2.5-mediated cardiovascular disease in aging: Cardiometabolic risks, molecular mechanisms and potential interventions. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176255. [PMID: 39276993 DOI: 10.1016/j.scitotenv.2024.176255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Air pollution, particularly fine particulate matter (PM2.5) with <2.5 μm in diameter, is a major public health concern. Studies have consistently linked PM2.5 exposure to a heightened risk of cardiovascular diseases (CVDs) such as ischemic heart disease (IHD), heart failure (HF), and cardiac arrhythmias. Notably, individuals with pre-existing age-related cardiometabolic conditions appear more susceptible. However, the specific impact of PM2.5 on CVDs susceptibility in older adults remains unclear. Therefore, this review addresses this gap by discussing the factors that make the elderly more vulnerable to PM2.5-induced CVDs. Accordingly, we focused on physiological aging, increased susceptibility, cardiometabolic risk factors, CVDs, and biological mechanisms. This review concludes by examining potential interventions to reduce exposure and the adverse health effects of PM2.5 in the elderly population. The latter includes dietary modifications, medications, and exploration of the potential benefits of supplements. By comprehensively analyzing these factors, this review aims to provide a deeper understanding of the detrimental effects of PM2.5 on cardiovascular health in older adults. This knowledge can inform future research and guide strategies to protect vulnerable populations from the adverse effects of air pollution.
Collapse
Affiliation(s)
- Francis Chanda
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Kai-Xuan Lin
- Department of Cardiology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China; Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Abdallah Iddy Chaurembo
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jian-Yuan Huang
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Hui-Juan Zhang
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Wen-Hui Deng
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yun-Jing Xu
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Li
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Li-Dan Fu
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Hao-Dong Cui
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Chi Shu
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; Food Science College, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Yang Chen
- University of Chinese Academy of Sciences, Beijing, China; Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
| | - Na Xing
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China.
| | - Han-Bin Lin
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, Guangdong, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
25
|
Han QH, Huang SM, Wu SS, Luo SS, Lou ZY, Li H, Yang YM, Zhang Q, Shao JM, Zhu LJ. Mapping the evolution of liver aging research: A bibliometric analysis. World J Gastroenterol 2024; 30:4461-4480. [PMID: 39534417 PMCID: PMC11551677 DOI: 10.3748/wjg.v30.i41.4461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/16/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND With the increasing of the global aging population, healthy aging and prevention of age-related diseases have become increasingly important. The liver, a vital organ involved in metabolism, detoxification, digestion, and immunity, holds a pivotal role in the aging process of organisms. Although extensive research on liver aging has been carried out, no bibliometric analysis has been conducted to evaluate the scientific progress in this area. AIM To analyze basic knowledge, development trends, and current research frontiers in the field via bibliometric methods. METHODS We conducted bibliometric analyses via a range of analytical tools including Python, the bibliometrix package in R, CiteSpace, and VOSviewer. We retrieved publication data on liver aging research from the Web of Science Core Collection Database. A scientific knowledge map was constructed to display the contributions from different authors, journals, countries, institutions, as well as patterns of co-occurrence keywords and co-cited references. Additionally, gene regulation pathways associated with liver aging were analyzed via the STRING database. RESULTS We identified 4288 articles on liver aging, authored by 24034 contributors from 4092 institutions across 85 countries. Notably, the years 1991 and 2020 presented significant bursts in publication output. The United States led in terms of publications (n = 1008, 25.1%), citations (n = 55205), and international collaborations (multiple country publications = 214). Keywords such as "lipid metabolism", "fatty liver disease", "inflammation", "liver fibrosis" and "target" were prominent, highlighting the current research hotspots. Notably, the top 64 genes, each of which appeared in at least 8 articles, were involved in pathways essential for cell survival and aging, including the phosphatidylinositol 3-kinase/protein kinase B, Forkhead box O and p53 signaling pathways. CONCLUSION This study highlights key areas of liver aging and offers a comprehensive overview of research trends, as well as insights into potential value for collaborative pursuits and clinical implementations.
Collapse
Affiliation(s)
- Qun-Hua Han
- Department of Geriatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Department of Pathology & Pathophysiology, Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang Province, China
| | - Shun-Mei Huang
- Department of Geriatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Sha-Sha Wu
- Department of Geriatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Department of Rehabilitation Medicine, First People’s Hospital of Wenling, Wenling 317500, Zhejiang Province, China
| | - Sui-Sui Luo
- Department of Geriatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Zhi-Yuan Lou
- Department of Pathology & Pathophysiology, Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang Province, China
| | - Hui Li
- Laboratory of Animal Research Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Yun-Mei Yang
- Department of Geriatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Qin Zhang
- Department of Geriatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Ji-Min Shao
- Department of Pathology & Pathophysiology, Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang Province, China
| | - Li-Jun Zhu
- Department of Geriatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
26
|
Deng D, Yang S, Yu X, Zhou R, Liu Y, Zhang H, Cui D, Feng X, Wu Y, Qi X, Su Z. Aging-induced short-chain acyl-CoA dehydrogenase promotes age-related hepatic steatosis by suppressing lipophagy. Aging Cell 2024; 23:e14256. [PMID: 38898632 PMCID: PMC11464120 DOI: 10.1111/acel.14256] [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: 01/29/2024] [Revised: 05/06/2024] [Accepted: 06/09/2024] [Indexed: 06/21/2024] Open
Abstract
Hepatic steatosis, the first step in the development of nonalcoholic fatty liver disease (NAFLD), is frequently observed in the aging population. However, the underlying molecular mechanism remains largely unknown. In this study, we first employed GSEA enrichment analysis to identify short-chain acyl-CoA dehydrogenase (SCAD), which participates in the mitochondrial β-oxidation of fatty acids and may be associated with hepatic steatosis in elderly individuals. Subsequently, we examined SCAD expression and hepatic triglyceride content in various aged humans and mice and found that triglycerides were markedly increased and that SCAD was upregulated in aged livers. Our further evidence in SCAD-ablated mice suggested that SCAD deletion was able to slow liver aging and ameliorate aging-associated fatty liver. Examination of the molecular pathways by which the deletion of SCAD attenuates steatosis revealed that the autophagic degradation of lipid droplets, which was not detected in elderly wild-type mice, was maintained in SCAD-deficient old mice. This was due to the decrease in the production of acetyl-coenzyme A (acetyl-CoA), which is abundant in the livers of old wild-type mice. In conclusion, our findings demonstrate that the suppression of SCAD may prevent age-associated hepatic steatosis by promoting lipophagy and that SCAD could be a promising therapeutic target for liver aging and associated steatosis.
Collapse
Affiliation(s)
- Dan Deng
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of BiotherapySichuan UniversityChengduChina
| | - Shanshan Yang
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of BiotherapySichuan UniversityChengduChina
| | - Xiaoqian Yu
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of BiotherapySichuan UniversityChengduChina
| | - Ruixue Zhou
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of BiotherapySichuan UniversityChengduChina
| | - Yin Liu
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of BiotherapySichuan UniversityChengduChina
| | - Hongmei Zhang
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of BiotherapySichuan UniversityChengduChina
| | - Daxin Cui
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of BiotherapySichuan UniversityChengduChina
| | - Xingrong Feng
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of BiotherapySichuan UniversityChengduChina
| | - Yanting Wu
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of BiotherapySichuan UniversityChengduChina
| | - Xiaocun Qi
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of BiotherapySichuan UniversityChengduChina
| | - Zhiguang Su
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, and State Key Laboratory of BiotherapySichuan UniversityChengduChina
| |
Collapse
|
27
|
Liu W, Yang X, Zhan T, Huang M, Tian X, Tian X, Huang X. Weight-adjusted waist index is positively and linearly associated with all-cause and cardiovascular mortality in metabolic dysfunction-associated steatotic liver disease: findings from NHANES 1999-2018. Front Endocrinol (Lausanne) 2024; 15:1457869. [PMID: 39403588 PMCID: PMC11471496 DOI: 10.3389/fendo.2024.1457869] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/13/2024] [Indexed: 01/03/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease. Body mass index (BMI) is the most used obesity index but has important limitations. The weight-adjusted waist index (WWI) is a novel obesity metric and accurately reflects body composition. We explored the association of WWI with all-cause and cardiovascular disease (CVD) mortality in MASLD. METHODS Adult participants with MASLD were included from NHANES 1999-2018. WWI was calculated by dividing the waist circumference (WC) by the square root of body weight. MASLD was diagnosed by the presence of hepatic steatosis and at least one cardiometabolic risk factor in the absence of other causes of steatosis. A fatty liver index ≥60 suggested the presence of hepatic steatosis. Mortality data was obtained by prospectively linking to the National Death Index. Multivariate Cox proportional hazards regression analyses were used to explore these associations and multiple adjustment models were constructed including crude, partial, and fully adjusted models. RESULTS After adjusting for all covariates including BMI, WWI remained positively and linearly associated with all-cause and CVD mortality in MASLD (hazard ratios [HR] 1.247 and 1.218, respectively). Higher WWI was associated with a significantly increased risk of mortality (both p for trend <0.05). There was an "obesity paradox" between BMI and all-cause mortality in MASLD, with significantly lower all-cause mortality in those with overweight/obesity compared to normal BMI (HR 0.625 and 0.596, respectively, p for trend = 0.024), and no association between BMI and CVD mortality. Interaction analyses indicated that these associations were influenced by several demographic variables and disease status. Time-dependent receiver operating characteristic curves indicated that the predictive value of WWI for mortality in MASLD was higher than that of BMI, WC, and waist-to-height ratio across all follow-up durations. CONCLUSIONS WWI was positively and linearly associated with all-cause and CVD mortality in MASLD, whereas BMI did not accurately reflect mortality risk. WWI provided the optimal predictive value for mortality compared to traditional obesity indicators. These findings emphasize the potential use of WWI as a novel obesity indicator for mortality risk assessment, stratification, and prevention in MASLD.
Collapse
Affiliation(s)
- Weijie Liu
- Department of Gastroenterology, Tongren Hospital of Wuhan University (WuHan Third Hospital), Wuhan, China
| | - Xiulin Yang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ting Zhan
- Department of Gastroenterology, Tongren Hospital of Wuhan University (WuHan Third Hospital), Wuhan, China
| | - Min Huang
- Department of Gastroenterology, Tongren Hospital of Wuhan University (WuHan Third Hospital), Wuhan, China
| | - Xiaorong Tian
- Department of Gastroenterology, Tongren Hospital of Wuhan University (WuHan Third Hospital), Wuhan, China
| | - Xia Tian
- Department of Gastroenterology, Tongren Hospital of Wuhan University (WuHan Third Hospital), Wuhan, China
| | - Xiaodong Huang
- Department of Gastroenterology, Tongren Hospital of Wuhan University (WuHan Third Hospital), Wuhan, China
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
28
|
Krüger P, Schroll M, Fenzl F, Lederer EM, Hartinger R, Arnold R, Cagla Togan D, Guo R, Liu S, Petry A, Görlach A, Djabali K. Inflammation and Fibrosis in Progeria: Organ-Specific Responses in an HGPS Mouse Model. Int J Mol Sci 2024; 25:9323. [PMID: 39273272 PMCID: PMC11395088 DOI: 10.3390/ijms25179323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is an extremely rare genetic disorder that causes accelerated aging, due to a pathogenic variant in the LMNA gene. This pathogenic results in the production of progerin, a defective protein that disrupts the nuclear lamina's structure. In our study, we conducted a histopathological analysis of various organs in the LmnaG609G/G609G mouse model, which is commonly used to study HGPS. The objective of this study was to show that progerin accumulation drives systemic but organ-specific tissue damage and accelerated aging phenotypes. Our findings show significant fibrosis, inflammation, and dysfunction in multiple organ systems, including the skin, cardiovascular system, muscles, lungs, liver, kidneys, spleen, thymus, and heart. Specifically, we observed severe vascular fibrosis, reduced muscle regeneration, lung tissue remodeling, depletion of fat in the liver, and disruptions in immune structures. These results underscore the systemic nature of the disease and suggest that chronic inflammation and fibrosis play crucial roles in the accelerated aging seen in HGPS. Additionally, our study highlights that each organ responds differently to the toxic effects of progerin, indicating that there are distinct mechanisms of tissue-specific damage.
Collapse
Affiliation(s)
- Peter Krüger
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Moritz Schroll
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Felix Fenzl
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Eva-Maria Lederer
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Ramona Hartinger
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Rouven Arnold
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Deniz Cagla Togan
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Runjia Guo
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Shiyu Liu
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| | - Andreas Petry
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital, Heart Diseases, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80636 Munich, Germany
| | - Agnes Görlach
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital, Heart Diseases, German Heart Center Munich, Technical University Munich, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, 80636 Munich, Germany
| | - Karima Djabali
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany
| |
Collapse
|
29
|
Gao XF, Ji BY, Zhang JJ, Wang Z, Jiang S, Hu JN, Gong XJ, Zhang JT, Tsopmejio ISN, Li W. Ginsenoside Rg2 Attenuates Aging-Induced Liver Injury via Inhibiting Caspase 8-Mediated Pyroptosis, Apoptosis and Modulating Gut Microbiota. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1427-1449. [PMID: 39192676 DOI: 10.1142/s0192415x24500563] [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: 08/29/2024]
Abstract
Aging is an irresistible natural law of the progressive decline of body molecules, organs, and overall function with the passage of time, resulting in eventual death. World Health Organization data show that aging is correlated with a wide range of common chronic diseases in the elderly, and is an essential driver of many diseases. Panax Ginseng C.A Meyer is an ancient herbal medicine, which has an effect of "long service, light weight, and longevity" recorded in the ancient Chinese medicine book "Compendium of Materia Medica." Ginsenoside Rg2, the main active ingredient of ginseng, also exerts a marked effect on the treatment of liver injury. However, it remains unclear whether Rg2 has the potential to ameliorate aging-induced liver injury. Hence, exploring the hepatoprotective properties of Rg2 and its possible molecular mechanism by Senescence Accelerate Mouse Prone 8 (SAMP8) and gut microbiota. Our study demonstrated that Rg2 can inhibit pyroptosis and apoptosis through caspase 8, and regulate the gut-liver axis to alleviate liver inflammation by changing the composition of gut microbiota, thus improving aging-induced liver injury. These findings provide theoretical support for the pharmacological effects of ginsenosides in delaying aging-induced liver injury.
Collapse
Affiliation(s)
- Xu-Fei Gao
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Bao-Yu Ji
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, P. R. China
- Tianjin Key Laboratory for Modern Drug Delivery and High Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Jun-Jie Zhang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Shuang Jiang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jun-Nan Hu
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Xiao-Jie Gong
- Department of Biological Engineering, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| | - Jing-Tian Zhang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Ivan Stève Nguepi Tsopmejio
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, P. R. China
| |
Collapse
|
30
|
Yin K, Büttner M, Deligiannis IK, Strzelecki M, Zhang L, Talavera-López C, Theis F, Odom DT, Martinez-Jimenez CP. Polyploidisation pleiotropically buffers ageing in hepatocytes. J Hepatol 2024; 81:289-302. [PMID: 38583492 DOI: 10.1016/j.jhep.2024.03.043] [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: 05/05/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND & AIMS Polyploidy in hepatocytes has been proposed as a genetic mechanism to buffer against transcriptional dysregulation. Here, we aim to demonstrate the role of polyploidy in modulating gene regulatory networks in hepatocytes during ageing. METHODS We performed single-nucleus RNA sequencing in hepatocyte nuclei of different ploidy levels isolated from young and old wild-type mice. Changes in the gene expression and regulatory network were compared to three independent strains that were haploinsufficient for HNF4A, CEBPA or CTCF, representing non-deleterious perturbations. Phenotypic characteristics of the liver section were additionally evaluated histologically, whereas the genomic allele composition of hepatocytes was analysed by BaseScope. RESULTS We observed that ageing in wild-type mice results in nuclei polyploidy and a marked increase in steatosis. Haploinsufficiency of liver-specific master regulators (HFN4A or CEBPA) results in the enrichment of hepatocytes with tetraploid nuclei at a young age, affecting the genomic regulatory network, and dramatically suppressing ageing-related steatosis tissue wide. Notably, these phenotypes are not the result of subtle disruption to liver-specific transcriptional networks, since haploinsufficiency in the CTCF insulator protein resulted in the same phenotype. Further quantification of genotypes of tetraploid hepatocytes in young and old HFN4A-haploinsufficient mice revealed that during ageing, tetraploid hepatocytes lead to the selection of wild-type alleles, restoring non-deleterious genetic perturbations. CONCLUSIONS Our results suggest a model whereby polyploidisation leads to fundamentally different cell states. Polyploid conversion enables pleiotropic buffering against age-related decline via non-random allelic segregation to restore a wild-type genome. IMPACT AND IMPLICATIONS The functional role of hepatocyte polyploidisation during ageing is poorly understood. Using single-nucleus RNA sequencing and BaseScope approaches, we have studied ploidy dynamics during ageing in murine livers with non-deleterious genetic perturbations. We have identified that hepatocytes present different cellular states and the ability to buffer ageing-associated dysfunctions. Tetraploid nuclei exhibit robust transcriptional networks and are better adapted to genomically overcome perturbations. Novel therapeutic interventions aimed at attenuating age-related changes in tissue function could be exploited by manipulation of ploidy dynamics during chronic liver conditions.
Collapse
Affiliation(s)
- Kelvin Yin
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany
| | - Maren Büttner
- Institute of Computational Biology, Computational Health Department, Helmholtz Munich, Neuherberg, Germany
| | | | | | - Liwei Zhang
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany
| | - Carlos Talavera-López
- Division of Infectious Diseases and Tropical Medicine, Ludwig-Maximilian-Universität Klinikum, Germany
| | - Fabian Theis
- Institute of Computational Biology, Computational Health Department, Helmholtz Munich, Neuherberg, Germany; Technical University of Munich, Department of Mathematics, 85748 Garching. Munich, Germany; German Cancer Research Centre, Heidelberg, Germany.
| | - Duncan T Odom
- German Cancer Research Center, Division of Regulatory Genomics and Cancer Evolution (B270), Heidelberg, Germany; Cancer Research UK Cambridge Institute, University of Cambridge, CB20RE, United Kingdom.
| | - Celia P Martinez-Jimenez
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany; TUM School of Medicine, Technical University of Munich, Munich, Germany; Institute of Biotechnology and Biomedicine (BIOTECMED), Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, Spain.
| |
Collapse
|
31
|
Dong W, Liu J, Zhang Y, Huang M, Lin M, Peng X. DNA damages in hepatocytes are amended by an inflammation-driven rescue repair mechanism in chronic hepatitis B. Pathol Res Pract 2024; 260:155391. [PMID: 38850878 DOI: 10.1016/j.prp.2024.155391] [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: 09/11/2023] [Revised: 04/23/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Our previous study has shown that intrahepatic necroinflammation favors the eliminations of HBV integration and clonal hepatocytes. Here, the effect of inflammation on host DNA damage eliminations in liver biopsy tissues from patients with chronic hepatitis B (CHB) was further investigated. METHODS DNA damage markers, histone γ-H2AX and phosphorylated heterochromatin protein 1γ (p-HP1γ), and senescent marker p21 were detected using immunohistochemical and immunofluorescent assays in liver biopsy samples from 69 CHB patients and 12 liver cirrhosis (LC) patients. Twenty paired hepatocellular carcinoma (HCC) surgical samples were used as controls. RESULTS Both γ-H2AX and p-HP1γ were sensitively detected in nuclear and cytoplasmic/nuclear patterns. Nuclear γ-H2AX was superior as a DNA damage marker in hepatocytes. The level of nuclear γ-H2AX in CHB, comparable to those in LC and HCC, was correlated with liver fibrosis and coexisted with the senescent marker p21. However, hepatocytes carried an alleviated level of DNA damages, which was associated with the level of cytoplasmic γ-H2AX. Cytoplasmic γ-H2AX chiefly occurred in hepatocytes near necroinflammatory foci, was correlated with liver inflammation and usually indicated the decrease or disappearance of nuclear γ-H2AX. The lack of cytoplasmic γ-H2AX together with the high level of nuclear γ-H2AX was associated with the progression from large cell changes/dysplasia to small cell changes/dysplasia. CONCLUSIONS Hepatocytes in CHB already carry massive DNA damages and undergo cellular senescence. The DNA damages in those senescent hepatocytes are histopathologically demonstrated to be amended by a novel cytoplasmic γ-H2AX-indicated and inflammation-driven rescue repair mechanism, which may be involved in hepatocarcinogenesis if it works improperly.
Collapse
Affiliation(s)
- Wenxiao Dong
- Department of Infectious Diseases, Jiangmen Central Hospital, Jiangmen, Guangdong 529000, China
| | - Jian Liu
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Yansong Zhang
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Mingxing Huang
- Department of Infectious Diseases, The Third People's Hospital of Zhuhai, Zhuhai, Guangdong 519000, China
| | - Minyi Lin
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China.
| | - Xiaomou Peng
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China.
| |
Collapse
|
32
|
Song CK, Ohlei O, Keller T, Regitz-Zagrosek V, Toepfer S, Steinhagen-Thiessen E, Bertram L, Buchmann N, Demuth I. Lipoprotein(a) and Lung Function Are Associated in Older Adults: Longitudinal and Cross-Sectional Analyses. Biomedicines 2024; 12:1502. [PMID: 39062075 PMCID: PMC11274407 DOI: 10.3390/biomedicines12071502] [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: 05/13/2024] [Revised: 06/12/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
While numerous studies have confirmed a causal association between lipoprotein(a) [Lp(a)] and cardiovascular diseases, only a few studies have assessed the relationship between Lp(a) and pulmonary health, with inconsistent findings regarding this topic. This study's aim was to examine whether levels of serum Lp(a) are associated with lung function in a dataset of relatively healthy older adults. We used longitudinal data collected at two time points 7.4 ± 1.5 years apart from 679 participants (52% women, 68 [65-71] years old) from the Berlin Aging Study II (BASE-II). Multiple linear regression models adjusting for covariates were applied to examine the association between Lp(a) and lung function. The forced expiratory volume in one second (FEV1) and the forced vital capacity (FVC) were higher in both men and women with higher Lp(a) levels. However, since this association between lung function parameters and Lp(a) was not supported by Mendelian randomization analyses using recent genome-wide association study data, these relationships should be investigated in future work, as the observed differences are, in part, considerable and potentially clinically relevant.
Collapse
Affiliation(s)
- Chae Kyung Song
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolic Diseases (Including Division of Lipid Metabolism), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Olena Ohlei
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, 23562 Lübeck, Germany
| | - Theresa Keller
- Institute of Biometry and Clinical Epidemiology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Reinhardtstraße 58, 10117 Berlin, Germany
| | - Vera Regitz-Zagrosek
- Institute for Gender in Medicine, Center for Cardiovascular Research, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 10099 Berlin, Germany
- Department of Cardiology, University Hospital Zürich, University of Zürich, 8057 Zürich, Switzerland
| | - Sarah Toepfer
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolic Diseases (Including Division of Lipid Metabolism), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Elisabeth Steinhagen-Thiessen
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolic Diseases (Including Division of Lipid Metabolism), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, 23562 Lübeck, Germany
| | - Nikolaus Buchmann
- Department of Cardiology, Charité–University Medicine Berlin, Campus Benjamin Franklin, 10117 Berlin, Germany
| | - Ilja Demuth
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolic Diseases (Including Division of Lipid Metabolism), Augustenburger Platz 1, 13353 Berlin, Germany
- Charité–Universitätsmedizin Berlin, BCRT-Berlin Institute of Health Center for Regenerative Therapies, 10117 Berlin, Germany
| |
Collapse
|
33
|
Vue Z, Murphy A, Le H, Neikirk K, Garza-Lopez E, Marshall AG, Mungai M, Jenkins B, Vang L, Beasley HK, Ezedimma M, Manus S, Whiteside A, Forni MF, Harris C, Crabtree A, Albritton CF, Jamison S, Demirci M, Prasad P, Oliver A, Actkins KV, Shao J, Zaganjor E, Scudese E, Rodriguez B, Koh A, Rabago I, Moore JE, Nguyen D, Aftab M, Kirk B, Li Y, Wandira N, Ahmad T, Saleem M, Kadam A, Katti P, Koh HJ, Evans C, Koo YD, Wang E, Smith Q, Tomar D, Williams CR, Sweetwyne MT, Quintana AM, Phillips MA, Hubert D, Kirabo A, Dash C, Jadiya P, Kinder A, Ajijola OA, Miller-Fleming TW, McReynolds MR, Hinton A. MICOS Complex Loss Governs Age-Associated Murine Mitochondrial Architecture and Metabolism in the Liver, While Sam50 Dictates Diet Changes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599846. [PMID: 38979162 PMCID: PMC11230271 DOI: 10.1101/2024.06.20.599846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The liver, the largest internal organ and a metabolic hub, undergoes significant declines due to aging, affecting mitochondrial function and increasing the risk of systemic liver diseases. How the mitochondrial three-dimensional (3D) structure changes in the liver across aging, and the biological mechanisms regulating such changes confers remain unclear. In this study, we employed Serial Block Face-Scanning Electron Microscopy (SBF-SEM) to achieve high-resolution 3D reconstructions of murine liver mitochondria to observe diverse phenotypes and structural alterations that occur with age, marked by a reduction in size and complexity. We also show concomitant metabolomic and lipidomic changes in aged samples. Aged human samples reflected altered disease risk. To find potential regulators of this change, we examined the Mitochondrial Contact Site and Cristae Organizing System (MICOS) complex, which plays a crucial role in maintaining mitochondrial architecture. We observe that the MICOS complex is lost during aging, but not Sam50. Sam50 is a component of the sorting and assembly machinery (SAM) complex that acts in tandem with the MICOS complex to modulate cristae morphology. In murine models subjected to a high-fat diet, there is a marked depletion of the mitochondrial protein SAM50. This reduction in Sam50 expression may heighten the susceptibility to liver disease, as our human biobank studies corroborate that Sam50 plays a genetically regulated role in the predisposition to multiple liver diseases. We further show that changes in mitochondrial calcium dysregulation and oxidative stress accompany the disruption of the MICOS complex. Together, we establish that a decrease in mitochondrial complexity and dysregulated metabolism occur with murine liver aging. While these changes are partially be regulated by age-related loss of the MICOS complex, the confluence of a murine high-fat diet can also cause loss of Sam50, which contributes to liver diseases. In summary, our study reveals potential regulators that affect age-related changes in mitochondrial structure and metabolism, which can be targeted in future therapeutic techniques.
Collapse
Affiliation(s)
- Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Alexandria Murphy
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Han Le
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Andrea G. Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Margaret Mungai
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Brenita Jenkins
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Mariaassumpta Ezedimma
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Sasha Manus
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Aaron Whiteside
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Maria Fernanda Forni
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520
| | - Chanel Harris
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208-3501, USA
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Claude F. Albritton
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208-3501, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sydney Jamison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mert Demirci
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Praveena Prasad
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Ashton Oliver
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ky’Era V. Actkins
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, 52242, USA
| | - Elma Zaganjor
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Estevão Scudese
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Benjamin Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Alice Koh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Izabella Rabago
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Johnathan E. Moore
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Desiree Nguyen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Muhammad Aftab
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Benjamin Kirk
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Yahang Li
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Nelson Wandira
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Taseer Ahmad
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Punjab,40100, Pakistan
| | - Mohammad Saleem
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ashlesha Kadam
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157 USA
| | - Prasanna Katti
- National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, AP, 517619, India
| | - Ho-Jin Koh
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Chantell Evans
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27708, USA
| | - Young Do Koo
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, Iowa, USA1
| | - Eric Wang
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, 92697, USA
| | - Quinton Smith
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, 92697, USA
| | - Dhanendra Tomar
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Punjab,40100, Pakistan
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435 USA
| | - Mariya T. Sweetwyne
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Anita M. Quintana
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, USA
| | - Mark A. Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, OR, 97331, USA
| | - David Hubert
- Department of Integrative Biology, Oregon State University, Corvallis, OR, 97331, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Nashville, TN, 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, 37232, USA
- Vanderbilt Institute for Global Health, Nashville, TN, 37232, USA
| | - Chandravanu Dash
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN, United States
| | - Pooja Jadiya
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, NC
| | - André Kinder
- Artur Sá Earp Neto University Center – UNIFASE-FMP, Petrópolis Medical School, Brazil
| | - Olujimi A. Ajijola
- UCLA Cardiac Arrhythmia Center, University of California, Los Angeles, CA, USA
| | - Tyne W. Miller-Fleming
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Melanie R. McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
34
|
Kirchner VA, Badshah JS, Kyun Hong S, Martinez O, Pruett TL, Niedernhofer LJ. Effect of Cellular Senescence in Disease Progression and Transplantation: Immune Cells and Solid Organs. Transplantation 2024; 108:1509-1523. [PMID: 37953486 PMCID: PMC11089077 DOI: 10.1097/tp.0000000000004838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Aging of the world population significantly impacts healthcare globally and specifically, the field of transplantation. Together with end-organ dysfunction and prolonged immunosuppression, age increases the frequency of comorbid chronic diseases in transplant candidates and recipients, contributing to inferior outcomes. Although the frequency of death increases with age, limited use of organs from older deceased donors reflects the concerns about organ durability and inadequate function. Cellular senescence (CS) is a hallmark of aging, which occurs in response to a myriad of cellular stressors, leading to activation of signaling cascades that stably arrest cell cycle progression to prevent tumorigenesis. In aging and chronic conditions, senescent cells accumulate as the immune system's ability to clear them wanes, which is causally implicated in the progression of chronic diseases, immune dysfunction, organ damage, decreased regenerative capacity, and aging itself. The intimate interplay between senescent cells, their proinflammatory secretome, and immune cells results in a positive feedback loop, propagating chronic sterile inflammation and the spread of CS. Hence, senescent cells in organs from older donors trigger the recipient's alloimmune response, resulting in the increased risk of graft loss. Eliminating senescent cells or attenuating their inflammatory phenotype is a novel, potential therapeutic target to improve transplant outcomes and expand utilization of organs from older donors. This review focuses on the current knowledge about the impact of CS on circulating immune cells in the context of organ damage and disease progression, discusses the impact of CS on abdominal solid organs that are commonly transplanted, and reviews emerging therapies that target CS.
Collapse
Affiliation(s)
- Varvara A. Kirchner
- Division of Abdominal Transplantation, Department of Surgery, Stanford University, Stanford, CA
| | - Joshua S. Badshah
- Division of Abdominal Transplantation, Department of Surgery, Stanford University, Stanford, CA
| | - Suk Kyun Hong
- Division of Abdominal Transplantation, Department of Surgery, Stanford University, Stanford, CA
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Olivia Martinez
- Division of Abdominal Transplantation, Department of Surgery, Stanford University, Stanford, CA
| | - Timothy L. Pruett
- Division of Transplantation, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN
| |
Collapse
|
35
|
Wang Y, Qiu H, Chen S, Li D, Zhao X, Guo M, Li N, Chen C, Qin M, Zhou Y, Xiao D, Zhao J, Xu L. MicroRNA-7 deficiency ameliorates d-galactose-induced aging in mice by regulating senescence of Kupffer cells. Aging Cell 2024; 23:e14145. [PMID: 38494663 PMCID: PMC11166366 DOI: 10.1111/acel.14145] [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: 10/09/2023] [Revised: 02/05/2024] [Accepted: 03/01/2024] [Indexed: 03/19/2024] Open
Abstract
Aging is intricately linked to immune system dysfunction. Recent studies have highlighted the biological function of microRNA-7 (miR-7) as a novel regulator of immune cell function and related diseases. However, the potential role of miR-7 in aging remains unexplored. Here, we investigated the contribution of miR-7 to d-gal-induced aging in mice, focusing on its regulation of senescent Kupffer cells. Our findings revealed that miR-7 deficiency significantly ameliorated the aging process, characterized by enhanced CD4+ T-cell activation. However, the adoptive transfer of miR-7-deficient CD4+T cells failed to improve the age-related phenotype. Further analysis showed that miR-7 deficiency significantly reduced IL-1β production in liver tissue, and inhibiting IL-1β in vivo slowed down the aging process in mice. Notably, IL-1β is mainly produced by senescent Kupffer cells in the liver tissue of aging mice, and miR-7 expression was significantly up-regulated in these cells. Mechanistically, KLF4, a target of miR-7, was down-regulated in senescent Kupffer cells in aging mouse model. Furthermore, miR-7 deficiency also modulated the NF-κB activation and IL-1β production in senescent Kupffer cells through KLF4. In conclusion, our findings unveil the role of miR-7 in d-gal-induced aging in mice, highlighting its regulation of KLF4/NF-κB/IL-1β pathways in senescent Kupffer cells. This research may enhance our understanding of miRNA-based aging immune cells and offer new avenues for new intervention strategies in aging process.
Collapse
Affiliation(s)
- Ya Wang
- Department of ImmunologyZunyi Medical UniversityZunyiGuizhouChina
- Key Laboratory of Gene Detection and Treatment of Guizhou ProvinceZunyiGuizhouChina
| | - Hui Qiu
- Department of ImmunologyZunyi Medical UniversityZunyiGuizhouChina
- Key Laboratory of Gene Detection and Treatment of Guizhou ProvinceZunyiGuizhouChina
| | - Shipeng Chen
- Department of ImmunologyZunyi Medical UniversityZunyiGuizhouChina
- Key Laboratory of Gene Detection and Treatment of Guizhou ProvinceZunyiGuizhouChina
| | - Dongmei Li
- Department of ImmunologyZunyi Medical UniversityZunyiGuizhouChina
- Key Laboratory of Gene Detection and Treatment of Guizhou ProvinceZunyiGuizhouChina
| | - Xu Zhao
- Key Laboratory of Gene Detection and Treatment of Guizhou ProvinceZunyiGuizhouChina
- School of MedicineGuizhou UniversityGuiyangGuizhouChina
| | - Mengmeng Guo
- Department of ImmunologyZunyi Medical UniversityZunyiGuizhouChina
- Key Laboratory of Gene Detection and Treatment of Guizhou ProvinceZunyiGuizhouChina
| | - Nana Li
- Key Laboratory of Gene Detection and Treatment of Guizhou ProvinceZunyiGuizhouChina
| | - Chao Chen
- Department of ImmunologyZunyi Medical UniversityZunyiGuizhouChina
- Key Laboratory of Gene Detection and Treatment of Guizhou ProvinceZunyiGuizhouChina
| | - Ming Qin
- Department of ImmunologyZunyi Medical UniversityZunyiGuizhouChina
- Key Laboratory of Gene Detection and Treatment of Guizhou ProvinceZunyiGuizhouChina
| | - Ya Zhou
- Department of Medical PhysicsZunyi Medical UniversityZunyiGuizhouChina
| | | | - Juanjuan Zhao
- Department of ImmunologyZunyi Medical UniversityZunyiGuizhouChina
- Key Laboratory of Gene Detection and Treatment of Guizhou ProvinceZunyiGuizhouChina
| | - Lin Xu
- Department of ImmunologyZunyi Medical UniversityZunyiGuizhouChina
- Key Laboratory of Gene Detection and Treatment of Guizhou ProvinceZunyiGuizhouChina
| |
Collapse
|
36
|
Nauffal V, Klarqvist MDR, Hill MC, Pace DF, Di Achille P, Choi SH, Rämö JT, Pirruccello JP, Singh P, Kany S, Hou C, Ng K, Philippakis AA, Batra P, Lubitz SA, Ellinor PT. Noninvasive assessment of organ-specific and shared pathways in multi-organ fibrosis using T1 mapping. Nat Med 2024; 30:1749-1760. [PMID: 38806679 DOI: 10.1038/s41591-024-03010-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/22/2024] [Indexed: 05/30/2024]
Abstract
Fibrotic diseases affect multiple organs and are associated with morbidity and mortality. To examine organ-specific and shared biologic mechanisms that underlie fibrosis in different organs, we developed machine learning models to quantify T1 time, a marker of interstitial fibrosis, in the liver, pancreas, heart and kidney among 43,881 UK Biobank participants who underwent magnetic resonance imaging. In phenome-wide association analyses, we demonstrate the association of increased organ-specific T1 time, reflecting increased interstitial fibrosis, with prevalent diseases across multiple organ systems. In genome-wide association analyses, we identified 27, 18, 11 and 10 independent genetic loci associated with liver, pancreas, myocardial and renal cortex T1 time, respectively. There was a modest genetic correlation between the examined organs. Several loci overlapped across the examined organs implicating genes involved in a myriad of biologic pathways including metal ion transport (SLC39A8, HFE and TMPRSS6), glucose metabolism (PCK2), blood group antigens (ABO and FUT2), immune function (BANK1 and PPP3CA), inflammation (NFKB1) and mitosis (CENPE). Finally, we found that an increasing number of organs with T1 time falling in the top quintile was associated with increased mortality in the population. Individuals with a high burden of fibrosis in ≥3 organs had a 3-fold increase in mortality compared to those with a low burden of fibrosis across all examined organs in multivariable-adjusted analysis (hazard ratio = 3.31, 95% confidence interval 1.77-6.19; P = 1.78 × 10-4). By leveraging machine learning to quantify T1 time across multiple organs at scale, we uncovered new organ-specific and shared biologic pathways underlying fibrosis that may provide therapeutic targets.
Collapse
Affiliation(s)
- Victor Nauffal
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Matthew C Hill
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Danielle F Pace
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paolo Di Achille
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joel T Rämö
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - James P Pirruccello
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| | - Pulkit Singh
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shinwan Kany
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, University Heart and Vascular Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cody Hou
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kenney Ng
- Center for Computational Health, IBM Research, Cambridge, MA, USA
| | - Anthony A Philippakis
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Eric and Wendy Schmidt Center, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Puneet Batra
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Steven A Lubitz
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
37
|
Šošić-Jurjević B, Borković-Mitić S, Pavlović S, Vlahović D, Miler M, Cesar T, Ajdžanović V, Milenkovic D, Stellaard F, Trifunović S, Filipović B, Lütjohann D. Lemon Flavonoid Extract Eriomin Improves Pro/Antioxidant Status and Interferes with Cholesterol Metabolism without Affecting Serum Cholesterol Levels in Aged Rats. Int J Mol Sci 2024; 25:5221. [PMID: 38791260 PMCID: PMC11121178 DOI: 10.3390/ijms25105221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/28/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
This study aimed to assess the antioxidant capacity of lemon flavonoid extract Eriomin® (LE) and its impact on cholesterol metabolism in the context of healthy aging. We orally treated 24-month-old male Wistar rats with an LE (40 mg/kg) suspended in 0.3 mL of sunflower oil. At the same time, control groups received an equal volume of sunflower oil (CON) or remained untreated (ICON) daily for 4 weeks. We examined LE's effects on superoxide dismutase and catalase- and glutathione-related enzyme activities, the concentration of lipid peroxides and protein carbonyls, total oxidant status (TOS) and antioxidant status (TAS), and oxidative stress index (OSI) in the liver, jejunum, and ileum. We also measured total cholesterol, its biosynthetic precursors (lanosterol, lathosterol, desmosterol), its degradation products (bile acid precursors) in the serum, liver, jejunum, and ileum, and serum phytosterols (intestinal absorption markers). LE reduced TOS, TAS, and OSI (p < 0.05) compared with control values, indicating its consistent antioxidant action in all examined organs. LE lowered hepatic desmosterol (p < 0.05) while also reducing 7α- and 24-hydroxycholesterol levels in the liver and ileum (p < 0.01). Serum cholesterol, hepatic gene expression, and the immunostaining intensity of CYP7A1 were unchanged. In conclusion, LE exerted non-enzymatic antioxidant effects and reduced cholesterol degradation, reducing its biosynthesis products, thereby maintaining serum cholesterol levels.
Collapse
Affiliation(s)
- Branka Šošić-Jurjević
- Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia; (S.B.-M.); (S.P.); (D.V.); (M.M.); (V.A.); (S.T.); (B.F.)
| | - Slavica Borković-Mitić
- Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia; (S.B.-M.); (S.P.); (D.V.); (M.M.); (V.A.); (S.T.); (B.F.)
| | - Slađan Pavlović
- Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia; (S.B.-M.); (S.P.); (D.V.); (M.M.); (V.A.); (S.T.); (B.F.)
| | - Dragana Vlahović
- Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia; (S.B.-M.); (S.P.); (D.V.); (M.M.); (V.A.); (S.T.); (B.F.)
| | - Marko Miler
- Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia; (S.B.-M.); (S.P.); (D.V.); (M.M.); (V.A.); (S.T.); (B.F.)
| | - Thais Cesar
- Graduate Program in Food, Nutrition and Food Engineering, Sao Paulo State University (UNESP), Araraquara 14800-060, Brazil;
| | - Vladimir Ajdžanović
- Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia; (S.B.-M.); (S.P.); (D.V.); (M.M.); (V.A.); (S.T.); (B.F.)
| | - Dragan Milenkovic
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA;
| | - Frans Stellaard
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; (F.S.); (D.L.)
| | - Svetlana Trifunović
- Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia; (S.B.-M.); (S.P.); (D.V.); (M.M.); (V.A.); (S.T.); (B.F.)
| | - Branko Filipović
- Institute for Biological Research “Siniša Stanković”—National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia; (S.B.-M.); (S.P.); (D.V.); (M.M.); (V.A.); (S.T.); (B.F.)
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; (F.S.); (D.L.)
| |
Collapse
|
38
|
Basudkar V, Gujrati G, Ajgaonkar S, Gandhi M, Mehta D, Nair S. Emerging Vistas for the Nutraceutical Withania somnifera in Inflammaging. Pharmaceuticals (Basel) 2024; 17:597. [PMID: 38794167 PMCID: PMC11123800 DOI: 10.3390/ph17050597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Inflammaging, a coexistence of inflammation and aging, is a persistent, systemic, low-grade inflammation seen in the geriatric population. Various natural compounds have been greatly explored for their potential role in preventing and treating inflammaging. Withania somnifera has been used for thousands of years in traditional medicine as a nutraceutical for its numerous health benefits including regenerative and adaptogenic effects. Recent preclinical and clinical studies on the role of Withania somnifera and its active compounds in treating aging, inflammation, and oxidative stress have shown promise for its use in healthy aging. We discuss the chemistry of Withania somnifera, the etiology of inflammaging and the protective role(s) of Withania somnifera in inflammaging in key organ systems including brain, lung, kidney, and liver as well as the mechanistic underpinning of these effects. Furthermore, we elucidate the beneficial effects of Withania somnifera in oxidative stress/DNA damage, immunomodulation, COVID-19, and the microbiome. We also delineate a putative protein-protein interaction network of key biomarkers modulated by Withania somnifera in inflammaging. In addition, we review the safety/potential toxicity of Withania somnifera as well as global clinical trials on Withania somnifera. Taken together, this is a synthetic review on the beneficial effects of Withania somnifera in inflammaging and highlights the potential of Withania somnifera in improving the health-related quality of life (HRQoL) in the aging population worldwide.
Collapse
Affiliation(s)
- Vivek Basudkar
- PhytoVeda Pvt. Ltd., Mumbai 400 022, India
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| | - Gunjan Gujrati
- PhytoVeda Pvt. Ltd., Mumbai 400 022, India
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| | - Saiprasad Ajgaonkar
- PhytoVeda Pvt. Ltd., Mumbai 400 022, India
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| | - Manav Gandhi
- College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Dilip Mehta
- PhytoVeda Pvt. Ltd., Mumbai 400 022, India
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| | - Sujit Nair
- PhytoVeda Pvt. Ltd., Mumbai 400 022, India
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| |
Collapse
|
39
|
Gieseler RK, Baars T, Özçürümez MK, Canbay A. Liver Diseases: Science, Fiction and the Foreseeable Future. J Pers Med 2024; 14:492. [PMID: 38793074 PMCID: PMC11122384 DOI: 10.3390/jpm14050492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
This Editorial precedes the Special Issue entitled "Novel Challenges and Therapeutic Options for Liver Diseases". Following a historical outline of the roots of hepatology, we provide a brief insight into our colleagues' contributions in this issue on the current developments in this discipline related to the prevention of liver diseases, the metabolic dysfunction-associated steatotic liver disease (or non-alcoholic fatty liver disease, respectively), liver cirrhosis, chronic viral hepatitides, acute-on-chronic liver failure, liver transplantation, the liver-microbiome axis and microbiome transplantation, and telemedicine. We further add some topics not covered by the contributions herein that will likely impact future hepatology. Clinically, these comprise the predictive potential of organokine crosstalk and treatment options for liver fibrosis. With regard to promising developments in basic research, some current findings on the genetic basis of metabolism-associated chronic liver diseases, chronobiology, metabolic zonation of the liver, aspects of the aging liver against the background of demography, and liver regeneration will be presented. We expect machine learning to thrive as an overarching topic throughout hepatology. The largest study to date on the early detection of liver damage-which has been kicked off on 1 March 2024-is highlighted, too.
Collapse
Affiliation(s)
- Robert K. Gieseler
- Department of Medicine, University Hospital Knappschaftskrankenhaus, Ruhr University Bochum, 44892 Bochum, Germany; (T.B.); (M.K.Ö.)
| | | | | | - Ali Canbay
- Department of Medicine, University Hospital Knappschaftskrankenhaus, Ruhr University Bochum, 44892 Bochum, Germany; (T.B.); (M.K.Ö.)
| |
Collapse
|
40
|
Chen X, Lu T, Zheng Y, Lin Z, Liu C, Yuan D, Yuan C. miR-155-5p promotes hepatic steatosis via PICALM-mediated autophagy in aging hepatocytes. Arch Gerontol Geriatr 2024; 120:105327. [PMID: 38237377 DOI: 10.1016/j.archger.2024.105327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 03/10/2024]
Abstract
BACKGROUND Hepatic steatosis, a lipid disorder characterized by the accumulation of intrahepatic fat, is more prevalent in the elderly population. This study investigates the role of miR-155-5p in the autophagy dysregulation of aging hepatic steatosis. METHODS We established an aging mouse model in vivo and a hepatocellular senescence model induced by low serum and palmitic acid in vitro. The fluctuations of microRNAs were derived from RNA-seq data and confirmed by qPCR in 4- and 18-month-old mouse liver tissues. Hematoxylin-eosin (H&E) staining observed pathological changes. Markers of senescence, autophagy, and lipolysis genes were analyzed using Western blot and qPCR. Bioinformatics analysis predicted miR-155-5p's target gene PICALM, confirmed by dual luciferase reporter assay and transfection of miR-155-5p mimic/inhibitor into senescent hepatocytes. RESULTS Senescent markers (p21, p16, and p-P53) and miR-155-5p were up-regulated in aging liver tissues and senescent hepatocytes. Bioinformatics analysis identified PICALM as a target gene of miR-155-5p, a finding further supported by dual luciferase reporter assays. Inhibition of miR-155-5p reduced expression of senescent marker genes (p16, p21, p-P53), improved autophagy (evidenced by increased LC3B-II and ATG5, and decreased P62), and enhanced lipolysis (indicated by increased ATGL and p-HSL) in senescent hepatocytes. Oil red O staining confirmed that miR-155-5p inhibition significantly reduced lipid accumulation in these cells. CONCLUSIONS This study suggests a potential new therapeutic approach for age-related hepatic steatosis through the inhibition of miR-155-5p to enhance autophagy.
Collapse
Affiliation(s)
- Xiaoling Chen
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China
| | - Ting Lu
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China
| | - Ying Zheng
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China
| | - Zhiyong Lin
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China
| | - Chaoqi Liu
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China.
| | - Ding Yuan
- College of Medicine and Health Science, China Three Gorges University, Yichang, HuBei, 443002, China.
| | - Chengfu Yuan
- College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China; Third Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, China Three Gorges University, School of Medicine, Yichang, 443002, China.
| |
Collapse
|
41
|
Kumar P, Hassan M, Tacke F, Engelmann C. Delineating the heterogeneity of senescence-induced-functional alterations in hepatocytes. Cell Mol Life Sci 2024; 81:200. [PMID: 38684535 PMCID: PMC11058795 DOI: 10.1007/s00018-024-05230-2] [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: 10/24/2023] [Revised: 03/11/2024] [Accepted: 04/07/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND AND AIM Cellular senescence of hepatocytes involves permanent cell cycle arrest, disrupted cellular bioenergetics, resistance to cell death, and the release of pro-inflammatory cytokines. This 'zombie-like' state perpetuates harmful effects on tissues and holds potential implications for liver disease progression. Remarkably, senescence exhibits heterogeneity, stemming from two crucial factors: the inducing stressor and the cell type. As such, our present study endeavors to characterize stressor-specific changes in senescence phenotype, its related molecular patterns, and cellular bioenergetics in primary mouse hepatocytes (PMH) and hepatocyte-derived liver organoids (HepOrgs). METHODS PMH, isolated by collagenase-perfused mouse liver (C57B6/J; 18-23 weeks), were cultured overnight in William's E-medium supplemented with 2% FBS, L-glutamine, and hepatocyte growth supplements. HepOrgs were developed by culturing cells in a 3D matrix for two weeks. The senescence was induced by DNA damage (doxorubicin, cisplatin, and etoposide), oxidative stress (H2O2, and ethanol), and telomere inhibition (BIBR-1532), p53 activation (nutlin-3a), DNA methyl transferase inhibition (5-azacitidine), and metabolism inhibitors (galactosamine and hydroxyurea). SA-β galactosidase activity, immunofluorescence, immunoblotting, and senescence-associated secretory phenotype (SASP), and cellular bioenergetics were used to assess the senescence phenotype. RESULTS Each senescence inducer triggers a unique combination of senescence markers in hepatocytes. All senescence inducers, except hydroxyurea and ethanol, increased SA-β galactosidase activity, the most commonly used marker for cellular senescence. Among the SASP factors, CCL2 and IL-10 were consistently upregulated, while Plasminogen activator inhibitor-1 exhibited global downregulation across all modes of senescence. Notably, DNA damage response was activated by DNA damage inducers. Cell cycle markers were most significantly reduced by doxorubicin, cisplatin, and galactosamine. Additionally, DNA damage-induced senescence shifted cellular bioenergetics capacity from glycolysis to oxidative phosphorylation. In HepOrgs exposed to senescence inducers, there was a notable increase in γH2A.X, p53, and p21 levels. Interestingly, while showing a similar trend, SASP gene expression in HepOrgs was significantly higher compared to PMH, demonstrating a several-fold increase. CONCLUSION In our study, we demonstrated that each senescence inducer activates a unique combination of senescence markers in PMH. Doxorubicin demonstrated the highest efficacy in inducing senescence, followed by cisplatin and H2O2, with no impact on apoptosis. Each inducer prompted DNA damage response and mitochondrial dysfunction, independent of MAPK/AKT.
Collapse
Affiliation(s)
- Pavitra Kumar
- Department of Hepatology and Gastroenterology, Medizinische Klinik m. S. Hepatologie und Gastroenterologie, Charité, Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Augustenburger Platz 1, Forum 4, Raum 2.0704a, 13353, Berlin, Germany
| | - Mohsin Hassan
- Department of Hepatology and Gastroenterology, Medizinische Klinik m. S. Hepatologie und Gastroenterologie, Charité, Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Augustenburger Platz 1, Forum 4, Raum 2.0704a, 13353, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Medizinische Klinik m. S. Hepatologie und Gastroenterologie, Charité, Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Augustenburger Platz 1, Forum 4, Raum 2.0704a, 13353, Berlin, Germany
| | - Cornelius Engelmann
- Department of Hepatology and Gastroenterology, Medizinische Klinik m. S. Hepatologie und Gastroenterologie, Charité, Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Augustenburger Platz 1, Forum 4, Raum 2.0704a, 13353, Berlin, Germany.
- Berlin Institute of Health (BIH), 10178, Berlin, Germany.
| |
Collapse
|
42
|
Hunt NJ, Lockwood GP, Heffernan SJ, Daymond J, Ngu M, Narayanan RK, Westwood LJ, Mohanty B, Esser L, Williams CC, Kuncic Z, McCourt PAG, Le Couteur DG, Cogger VC. Oral nanotherapeutic formulation of insulin with reduced episodes of hypoglycaemia. NATURE NANOTECHNOLOGY 2024; 19:534-544. [PMID: 38168926 PMCID: PMC11026164 DOI: 10.1038/s41565-023-01565-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 11/02/2023] [Indexed: 01/05/2024]
Abstract
Injectable insulin is an extensively used medication with potential life-threatening hypoglycaemic events. Here we report on insulin-conjugated silver sulfide quantum dots coated with a chitosan/glucose polymer to produce a responsive oral insulin nanoformulation. This formulation is pH responsive, is insoluble in acidic environments and shows increased absorption in human duodenum explants and Caenorhabditis elegans at neutral pH. The formulation is sensitive to glucosidase enzymes to trigger insulin release. It is found that the formulation distributes to the liver in mice and rats after oral administration and promotes a dose-dependent reduction in blood glucose without promoting hypoglycaemia or weight gain in diabetic rodents. Non-diabetic baboons also show a dose-dependent reduction in blood glucose. No biochemical or haematological toxicity or adverse events were observed in mice, rats and non-human primates. The formulation demonstrates the potential to orally control blood glucose without hypoglycaemic episodes.
Collapse
Affiliation(s)
- Nicholas J Hunt
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia.
- Sydney Nano Institute, The University of Sydney, Camperdown, New South Wales, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia.
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia.
| | - Glen P Lockwood
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia
| | - Scott J Heffernan
- Royal Prince Alfred Hospital, SLHD, Camperdown, New South Wales, Australia
| | - Jarryd Daymond
- Sydney Nano Institute, The University of Sydney, Camperdown, New South Wales, Australia
- Sydney Business School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Meng Ngu
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia
- Department of Gastroenterology, Concord Repatriation General Hospital, SLHD, Concord, New South Wales, Australia
| | - Ramesh K Narayanan
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia
| | - Lara J Westwood
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Sydney Nano Institute, The University of Sydney, Camperdown, New South Wales, Australia
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia
| | - Biswaranjan Mohanty
- Sydney Analytical Core Research Facility, The University of Sydney, Camperdown, New South Wales, Australia
| | - Lars Esser
- CSIRO Manufacturing, Clayton, Victoria, Australia
| | | | - Zdenka Kuncic
- Sydney Nano Institute, The University of Sydney, Camperdown, New South Wales, Australia
- School of Physics, The University of Sydney, Camperdown, New South Wales, Australia
| | - Peter A G McCourt
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia
- Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - David G Le Couteur
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia
| | - Victoria C Cogger
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia.
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia.
| |
Collapse
|
43
|
Zhang K, Jiang L, Xue L, Wang Y, Sun Y, Fan M, Qian H, Wang L, Li Y. 5-Heptadecylresorcinol Improves Aging-Associated Hepatic Fatty Acid Oxidation Dysfunction via Regulating Adipose Sirtuin 3. Nutrients 2024; 16:978. [PMID: 38613012 PMCID: PMC11013747 DOI: 10.3390/nu16070978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Aging-associated hepatic fatty acid (FA) oxidation dysfunction contributes to impaired adaptive thermogenesis. 5-Heptadecylresorcinol (AR-C17) is a prominent functional component of whole wheat and rye, and has been demonstrated to improve the thermogenic capacity of aged mice via the regulation of Sirt3. However, the effect of AR-C17 on aging-associated hepatic FA oxidation dysfunction remains unclear. Here, 18-month-old C57BL/6J mice were orally administered with AR-C17 at a dose of 150 mg/kg/day for 8 weeks. Systemic glucose and lipid metabolism, hepatic FA oxidation, and the lipolysis of white adipose tissues (WAT) were measured. The results showed that AR-C17 improved the hepatic FA oxidation, and especially acylcarnitine metabolism, of aged mice during cold stimulation, with the enhancement of systemic glucose and lipid metabolism. Meanwhile, AR-C17 improved the WAT lipolysis of aged mice, promoting hepatic acylcarnitine production. Furthermore, the adipose-specific Sirt3 knockout mice were used to investigate and verify the regulation mechanism of AR-C17 on aging-associated hepatic FA oxidation dysfunction. The results showed that AR-C17 failed to improve the WAT lipolysis and hepatic FA oxidation of aged mice in the absence of adipose Sirt3, indicating that AR-C17 might indirectly influence hepatic FA oxidation via regulating WAT Sirt3. Our findings suggest that AR-C17 might improve aging-associated hepatic FA oxidation dysfunction via regulating adipose Sirt3.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Li Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (K.Z.); (L.J.); (L.X.); (Y.W.); (Y.S.); (M.F.); (H.Q.)
| | - Yan Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (K.Z.); (L.J.); (L.X.); (Y.W.); (Y.S.); (M.F.); (H.Q.)
| |
Collapse
|
44
|
Wang Z, Cui J, Li X, Gao R, Feng E, Luo G, Guo B, Wu H, Sun Y, Sun J. Nomogram for predicting the risk of nonalcoholic fatty liver disease in older adults in Qingdao, China: A cross-sectional study. Asia Pac J Clin Nutr 2024; 33:83-93. [PMID: 38494690 PMCID: PMC11170012 DOI: 10.6133/apjcn.202403_33(1).0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/20/2023] [Accepted: 11/13/2023] [Indexed: 03/19/2024]
Abstract
BACKGROUND AND OBJECTIVES To explore the risk factors for non-alcoholic fatty liver disease (NAFLD) and to establish a non-invasive tool for the screening of NAFLD in an older adult population. METHODS AND STUDY DESIGN A total of 131,161 participants were included in this cross-sectional study. Participants were randomly divided into training and validation sets (7:3). The least absolute shrinkage and selection operator method was used to screen risk factors. Multivariate logistic regression was employed to develop a nomogram, which was made available online. Receiver operating characteristic curve analysis, calibration plots, and decision curve analysis were used to validate the discrimination, calibration, and clinical practicability of the nomogram. Sex and age subgroup analyses were conducted to further validate the reliability of the model. RESULTS Nine variables were identified for inclusion in the nomogram (age, sex, waist circumference, body mass index, exercise frequency, systolic blood pressure, fasting plasma glucose, alanine aminotransferase, and low-density lipoprotein cholesterol). The area under the receiver operating characteristic curve values were 0.793 and 0.790 for the training set and the validation set, respectively. The calibration plots and decision curve analyses showed good calibration and clinical utility. Subgroup analyses demonstrated consistent discriminatory ability in different sex and age subgroups. CONCLUSIONS This study established and validated a new nomogram model for evaluating the risk of NAFLD among older adults. The nomogram had good discriminatory performance and is a non-invasive and convenient tool for the screening of NAFLD in older adults.
Collapse
Affiliation(s)
- Zhi Wang
- School of Public Health, Qingdao University, Qingdao, China
| | - Jing Cui
- Qingdao Centers for Disease Control and Prevention/Qingdao Institute for Preventive Medicine, Qingdao China
| | - Xiaojing Li
- Qingdao Centers for Disease Control and Prevention/Qingdao Institute for Preventive Medicine, Qingdao China
| | - Ruili Gao
- Anqiu People's Hospital, Weifang, China
| | - Enqiang Feng
- Qingdao Centers for Disease Control and Prevention/Qingdao Institute for Preventive Medicine, Qingdao China
| | - Guoqiang Luo
- Qingdao Centers for Disease Control and Prevention/Qingdao Institute for Preventive Medicine, Qingdao China
| | - Baozhu Guo
- School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Haojia Wu
- School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Yongye Sun
- School of Public Health, Qingdao University, Qingdao, China.
| | - Jianping Sun
- Qingdao Centers for Disease Control and Prevention/Qingdao Institute for Preventive Medicine, Qingdao China.
| |
Collapse
|
45
|
Lin Y, Li Y, Liang G, Yang X, Yang J, Hu Q, Sun J, Zhang C, Fang H, Liu A. Single-cell transcriptome analysis of aging mouse liver. FASEB J 2024; 38:e23473. [PMID: 38334462 DOI: 10.1096/fj.202302282r] [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: 11/05/2023] [Revised: 12/30/2023] [Accepted: 01/24/2024] [Indexed: 02/10/2024]
Abstract
Aging has a great impact on the liver, which causes a loss of physiological integrity and an increase in susceptibility to injury, but many of the underlying molecular and cellular processes remain unclear. Here, we performed a comprehensive single-cell transcriptional profiling of the liver during aging. Our data showed that aging affected the cellular composition of the liver. The increase in inflammatory cells including neutrophils and monocyte-derived macrophages, as well as in inflammatory cytokines, could indicate an inflammatory tissue microenvironment in aged livers. Moreover, aging drove a distinct transcriptional course in each cell type. The commonly significant up-regulated genes were S100a8, S100a9, and RNA-binding motif protein 3 across all cell types. Aging-related pathways such as biosynthesis, metabolism, and oxidative stress were up-regulated in aged livers. Additionally, key ligand-receptor pairs for intercellular communication, primarily linked to macrophage migration inhibitory factor, transforming growth factor-β, and complement signaling, were also elevated. Furthermore, hepatic stellate cells (HSCs) serve as the prominent hub for intrahepatic signaling. HSCs acquired an "activated" phenotype, which may be involved in the increased intrahepatic vascular tone and fibrosis with aging. Liver sinusoidal endothelial cells derived from aged livers were pseudocapillarized and procontractile, and exhibited down-regulation of genes involved in vascular development and homeostasis. Moreover, the aging-related changes in cellular composition and gene expression were reversed by caloric restriction. Collectively, the present study suggests liver aging is linked to a significant liver sinusoidal deregulation and a moderate pro-inflammatory state, providing a potential concept for understanding the mechanism of liver aging.
Collapse
Affiliation(s)
- Yan Lin
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Ying Li
- Wuhan Fourth Hospital, Wuhan, China
| | - Guangyu Liang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Xiao Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Jiankun Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Qi Hu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Sun
- Department of Biliopancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cuntai Zhang
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoshu Fang
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| |
Collapse
|
46
|
Li S, Guo L. The role of Sirtuin 2 in liver - An extensive and complex biological process. Life Sci 2024; 339:122431. [PMID: 38242495 DOI: 10.1016/j.lfs.2024.122431] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 01/04/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
Liver disease has become one of the main causes of health issue worldwide. Sirtuin (Sirt) 2 is a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase, and is expressed in multiple organs including liver, which plays important and complex roles by interacting with various substrates. Physiologically, Sirt2 can improve metabolic homeostasis. Pathologically, Sirt2 can alleviate inflammation, endoplasmic reticulum (ER) stress, promote liver regeneration, maintain iron homeostasis, aggravate fibrogenesis and regulate oxidative stress in liver. In liver diseases, Sirt2 can mitigate fatty liver disease (FLD) including non-alcoholic fatty liver disease (NAFLD) and alcoholic fatty liver disease (AFLD), but aggravate hepatitis B (HBV) and liver ischemia-reperfusion injury (LIRI). The role of Sirt2 in liver cancer and aging-related liver diseases, however, has not been fully elucidated. In this review, these biological processes regulated by Sirt2 in liver are summarized, which aims to update the function of Sirt2 in liver and to explore the potential role of Sirt2 as a therapeutic target for liver diseases.
Collapse
Affiliation(s)
- Shan Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences (Shanghai University of Sport), Ministry of Education, Shanghai 200438, China
| | - Liang Guo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences (Shanghai University of Sport), Ministry of Education, Shanghai 200438, China.
| |
Collapse
|
47
|
Tran K, Gilbert M, Vazquez BN, Ianni A, Garcia BA, Vaquero A, Berger S. SIRT7 regulates NUCKS1 chromatin binding to elicit metabolic and inflammatory gene expression in senescence and liver aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.578810. [PMID: 38370824 PMCID: PMC10871251 DOI: 10.1101/2024.02.05.578810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Sirtuins, a class of highly conserved histone/protein deacetylases, are heavily implicated in senescence and aging. The regulation of sirtuin proteins is tightly controlled both transcriptionally and translationally and via localization within the cell. While Sirtiun proteins are implicated with aging, how their levels are regulated during aging across cell types and eliciting tissue specific age-related cellular changes is unclear. Here, we demonstrate that SIRT7 is targeted for degradation during senescence and liver aging. To uncover the significance of SIRT7 loss, we performed proteomics analysis and identified a new SIRT7 interactor, the HMG box protein NUCKS1. We found that the NUCKS1 transcription factor is recruited onto chromatin during senescence and this is mediated by SIRT7 loss. Further, depletion of NUCKS1 delayed senescence upon DNA damage leading to reduction of inflammatory gene expression. Examination of NUCKS1 transcriptional regulation during senescence revealed gene targets of transcription factors NFKB1, RELA, and CEBPβ. Consistently, in both Sirt7 KO mouse liver and in naturally aged livers, Nucks1 was recruited to chromatin. Further, Nucks1 was bound at promoters and enhancers of age-related genes, including transcription factor Rela, and, moreover, these bound sites had increased accessibility during aging. Overall, our results uncover NUCKS1 as a novel interactor of SIRT7, and show that loss of SIRT7 during senescence and liver aging promotes NUCKS1 chromatin binding to regulate metabolic and inflammatory genes.
Collapse
|
48
|
Draghi S, Curone G, Pavlovic R, Di Cesare F, Cagnardi P, Fornesi Silva C, Pellegrini A, Riva F, Arioli F, Fidani M. Influence of Area, Age and Sex on Per- and Polyfluorinated Alkyl Substances Detected in Roe Deer Muscle and Liver from Selected Areas of Northern Italy. Animals (Basel) 2024; 14:529. [PMID: 38396499 PMCID: PMC10885967 DOI: 10.3390/ani14040529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Due to their physicochemical properties, per- and polyfluorinated alkyl substances (PFASs) persist and bioaccumulate in living organisms, causing adverse health effects. Since exposure to xenobiotics is influenced by factors related to both the living organism and the considered compounds, biomonitoring PFASs' presence in the environment is of crucial importance. This study aimed to detect and quantify 15 PFASs in the muscle and liver of 40 roe deer from a specific area in Northern Italy by UPLC-HRMS. In the roe deer, liver PFAS concentrations were higher than those seen in muscle (p < 0.05). Although PFAS content in animals from urbanized areas was higher than those found in deer from rural areas, this difference was not statistically significant. In female roe deer, the concentration was higher than in males (p < 0.05); moreover, older animals showed higher concentrations of PFASs in the liver than younger animals (p < 0.05). In conclusion, the amount of PFASs was higher in tissues from roe deer belonging to urbanized areas, showing that this species might serve as a good bioindicator due to its territorial behavior. PFAS content was significantly higher in female roe deer, although the reason is not fully known. Finally, PFAS concentration was higher in the liver of older animals, probably due to compromised hepatic function.
Collapse
Affiliation(s)
- Susanna Draghi
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell’Università 6, 26900 Lodi, Italy; (S.D.); (G.C.); (P.C.); (F.R.); (F.A.)
| | - Giulio Curone
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell’Università 6, 26900 Lodi, Italy; (S.D.); (G.C.); (P.C.); (F.R.); (F.A.)
| | - Radmila Pavlovic
- Proteomics and Metabolomics Facility (ProMeFa), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy;
| | - Federica Di Cesare
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell’Università 6, 26900 Lodi, Italy; (S.D.); (G.C.); (P.C.); (F.R.); (F.A.)
| | - Petra Cagnardi
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell’Università 6, 26900 Lodi, Italy; (S.D.); (G.C.); (P.C.); (F.R.); (F.A.)
| | - Claudia Fornesi Silva
- UNIRELAB S.r.l., Via Gramsci 70, 20019 Settimo Milanese, Italy; (C.F.S.); (A.P.); (M.F.)
| | - Alberto Pellegrini
- UNIRELAB S.r.l., Via Gramsci 70, 20019 Settimo Milanese, Italy; (C.F.S.); (A.P.); (M.F.)
| | - Federica Riva
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell’Università 6, 26900 Lodi, Italy; (S.D.); (G.C.); (P.C.); (F.R.); (F.A.)
| | - Francesco Arioli
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Via dell’Università 6, 26900 Lodi, Italy; (S.D.); (G.C.); (P.C.); (F.R.); (F.A.)
| | - Marco Fidani
- UNIRELAB S.r.l., Via Gramsci 70, 20019 Settimo Milanese, Italy; (C.F.S.); (A.P.); (M.F.)
| |
Collapse
|
49
|
Naiini MR, Shahouzehi B, Azizi S, Shafiei B, Nazari-Robati M. Trehalose-induced SIRT1/AMPK activation regulates SREBP-1c/PPAR-α to alleviate lipid accumulation in aged liver. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1061-1070. [PMID: 37581638 DOI: 10.1007/s00210-023-02644-w] [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: 04/28/2023] [Accepted: 07/24/2023] [Indexed: 08/16/2023]
Abstract
Aging is associated with a disturbance in the regulation of the metabolic function of the liver, which increases the risk of liver and systemic diseases. Trehalose, a natural disaccharide, has been identified to reduce dyslipidemia, hepatic steatosis, and glucose intolerance. However, the roles of trehalose on lipid metabolism in aged liver are unclear which was investigated in this study. Thirty-two male Wistar rats were randomly allocated into four groups (n = 8). Two groups of aged (24 months) and young (4 months) rats were administered 2% trehalose solution orally for 30 days. Control groups of aged and young rats did not receive any treatment. At the end of the treatment period, blood samples and liver tissues were collected. Then the expression of SIRT1, AMPK, SREBP-1c, and PPAR-α and the level of AMPK phosphorylation (p-AMPK) were quantified by real-time polymerase chain reaction and western blotting. Moreover, biochemical parameters and the histopathology of livers were evaluated. Trehalose supplementation increased the level of SIRT1, p-AMPK, and PPAR-α, whereas the level of SREBP-1c was diminished in the liver of old animals. In addition, treatment with trehalose improved histopathological features of senescent livers. Taken together, our results show that old rats developed lipogenesis in the liver which was alleviated with trehalose. Therefore, trehalose may be an effective intervention to reduce the progression of aging-induced liver diseases.
Collapse
Affiliation(s)
- Mahdis Rahimi Naiini
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Beydolah Shahouzehi
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahrzad Azizi
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Bentolhoda Shafiei
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdieh Nazari-Robati
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
50
|
Jiang M, Zheng Z, Wang X, Chen Y, Qu J, Ding Q, Zhang W, Liu YS, Yang J, Tang W, Hou Y, He J, Wang L, Huang P, Li LC, He Z, Gao Q, Lu Q, Wei L, Wang YJ, Ju Z, Fan JG, Ruan XZ, Guan Y, Liu GH, Pei G, Li J, Wang Y. A biomarker framework for liver aging: the Aging Biomarker Consortium consensus statement. LIFE MEDICINE 2024; 3:lnae004. [PMID: 39872390 PMCID: PMC11749002 DOI: 10.1093/lifemedi/lnae004] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/29/2024] [Indexed: 01/11/2025]
Abstract
In human aging, liver aging per se not only increases susceptibility to liver diseases but also increases vulnerability of other organs given its central role in regulating metabolism. Total liver function tends to be well maintained in the healthy elderly, so liver aging is generally difficult to identify early. In response to this critical challenge, the Aging Biomarker Consortium of China has formulated an expert consensus on biomarkers of liver aging by synthesizing the latest scientific literature, comprising insights from both scientists and clinicians. This consensus provides a comprehensive assessment of biomarkers associated with liver aging and presents a systematic framework to characterize these into three dimensions: functional, imaging, and humoral. For the functional domain, we highlight biomarkers associated with cholesterol metabolism and liver-related coagulation function. For the imaging domain, we note that hepatic steatosis and liver blood flow can serve as measurable biomarkers for liver aging. Finally, in the humoral domain, we pinpoint hepatokines and enzymatic alterations worthy of attention. The aim of this expert consensus is to establish a foundation for assessing the extent of liver aging and identify early signs of liver aging-related diseases, thereby improving liver health and the healthy life expectancy of the elderly population.
Collapse
Affiliation(s)
| | - Mengmeng Jiang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhuozhao Zheng
- Department of Radiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Xuan Wang
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yanhao Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - You-Shuo Liu
- Department of Geriatrics, the Second Xiangya Hospital, and the Institute of Aging and Geriatrics, Central South University, Changsha 410011, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Weiqing Tang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Yunlong Hou
- Yiling Pharmaceutical Academician Workstation, Shijiazhuang 050035, China
| | - Jinhan He
- Department of Pharmacy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
| | - Pengyu Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Engineering Research Center of Pulmonary and Critical Care Medicine Technology and Device (Ministry of Education), Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Lin-Chen Li
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Zhiying He
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200092, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qian Lu
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education), School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yan-Jiang Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou 510632, China
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiong Zhong Ruan
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gang Pei
- Collaborative Innovation Center for Brain Science, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Yunfang Wang
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education), School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 102218, China
| |
Collapse
|