1
|
Fernández-Ramos D, Lopitz-Otsoa F, Lu SC, Mato JM. S-Adenosylmethionine: A Multifaceted Regulator in Cancer Pathogenesis and Therapy. Cancers (Basel) 2025; 17:535. [PMID: 39941901 PMCID: PMC11816870 DOI: 10.3390/cancers17030535] [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: 12/16/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
S-adenosylmethionine (SAMe) is a key methyl donor that plays a critical role in a variety of cellular processes, such as DNA, RNA and protein methylation, essential for maintaining genomic stability, regulating gene expression and maintaining cellular homeostasis. The involvement of SAMe in cancer pathogenesis is multifaceted, as through its multiple cellular functions, it can influence tumor initiation, progression and therapeutic resistance. In addition, the connection of SAMe with polyamine synthesis and oxidative stress management further underscores its importance in cancer biology. Recent studies have highlighted the potential of SAMe as a biomarker for cancer diagnosis and prognosis. Furthermore, the therapeutic implications of SAMe are promising, with evidence suggesting that SAMe supplementation or modulation could improve the efficacy of existing cancer treatments by restoring proper methylation patterns and mitigating oxidative damage and protect against damage induced by chemotherapeutic drugs. Moreover, targeting methionine cycle enzymes to both regulate SAMe availability and SAMe-independent regulatory effects, particularly in methionine-dependent cancers such as colorectal and lung cancer, presents a promising therapeutic approach. Additionally, exploring epitranscriptomic regulations, such as m6A modifications, and their interaction with non-coding RNAs could enhance our understanding of tumor progression and resistance mechanisms. Precision medicine approaches integrating patient subtyping and combination therapies with chemotherapeutics, such as decitabine or doxorubicin, together with SAMe, can enhance chemosensitivity and modulate epigenomics, showing promising results that may improve treatment outcomes. This review comprehensively examines the various roles of SAMe in cancer pathogenesis, its potential as a diagnostic and prognostic marker, and its emerging therapeutic applications. While SAMe modulation holds significant promise, challenges such as bioavailability, patient stratification and context-dependent effects must be addressed before clinical implementation. In addition, better validation of the obtained results into specific cancer animal models would also help to bridge the gap between research and clinical practice.
Collapse
Affiliation(s)
- David Fernández-Ramos
- Precision Medicine and Metabolism Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (D.F.-R.); (F.L.-O.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Fernando Lopitz-Otsoa
- Precision Medicine and Metabolism Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (D.F.-R.); (F.L.-O.)
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - José M. Mato
- Precision Medicine and Metabolism Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain; (D.F.-R.); (F.L.-O.)
| |
Collapse
|
2
|
Wang Q, Cheng W, He T, Li S, Ao J, He Y, Duan C, Li X, Zhang J. Glycerophospholipid metabolic disorders and gender difference of cantharidin-induced hepatotoxicity in rats: Lipidomics and MALDI mass spectrometry imaging analysis. Chem Biol Interact 2025; 405:111314. [PMID: 39551422 DOI: 10.1016/j.cbi.2024.111314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/14/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
The hepatotoxicity mechanism of cantharidin (CTD), a major active component of Mylabris was explored based on liver lipidome alterations and spatial distributions in female and male rats using lipidomics and matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI). After oral CTD exposure, the livers of female rats were screened for 104 differential lipids including lysophosphatidylethanolamine(LysoPE)(20:2/0:0) and diacylglycerol(DG)(18:2/22:4), whereas the livers of male rats were screened for 76 differential lipids including fatty acid(FA)(24:6) and DG(18:0/22:4). According to the MALDI-MSI results, female rats exhibited 12 differential lipids with alteration in the abundance and spatial distribution of phosphatylcholine(PC), phosphatidylethanolamine(PE), lysophosphatidylcholine(LysoPC), and LysoPE in the liver lesion area. On the other hand, male rats exhibited 8 differential lipids with changes in the abundance and spatial distribution of PC, PE, and FA in the liver lesion area. The lipidomics- and MALDI-MSI-detected differential lipids strongly disrupted glycerophospholipid metabolism in both female and male rats. Additionally, phosphatidate phosphatase (Lipin1), choline/ethanolamine phosphotransferase 1 (CEPT1), and phosphatidylethanolamine N-methyltransferase (PEMT) were screened to distinguish CTD hepatoxicity in female and male rats. Western blotting analysis demonstrated a significant elevation in Lipin1 expression in female and male rat livers, accompanied by a decrease in PEMT expression. Furthermore, CEPT1 expression increased significantly in female rat livers and decreased significantly in male rat livers. These findings suggested that CTD could disrupt lipid metabolism in a gender-specific manner. Moreover, the combination of lipidomics and MALDI-MSI could offer valuable insights into CTD-induced hepatotoxicity in rats.
Collapse
Affiliation(s)
- Qiyi Wang
- Department of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
| | - Weina Cheng
- Department of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
| | - Tianmu He
- Department of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
| | - Shan Li
- Department of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
| | - Jingwen Ao
- Department of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
| | - Yanmei He
- Department of Pharmacy, Zunyi Medical University, Zunyi, 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
| | - Cancan Duan
- Department of Pharmacy, Zunyi Medical University, Zunyi, 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
| | - Xiaofei Li
- School of Basic Medicine, Zunyi Medical University, Zunyi, 563000, China
| | - Jianyong Zhang
- Department of Pharmacy, Zunyi Medical University, Zunyi, 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
3
|
Tan EY, Muthiah MD, Sanyal AJ. Metabolomics at the cutting edge of risk prediction of MASLD. Cell Rep Med 2024; 5:101853. [PMID: 39657668 PMCID: PMC11722125 DOI: 10.1016/j.xcrm.2024.101853] [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: 07/25/2024] [Revised: 10/12/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major public health threat globally. Management of patients afflicted with MASLD and research in this domain are limited by the lack of robust well-established non-invasive biomarkers for diagnosis, prognostication, and monitoring. The circulating metabolome reflects both the systemic metabo-inflammatory milieu and changes in the liver in affected individuals. In this review we summarize the available literature on changes in the different components of the metabolome in MASLD with a focus on changes that are linked to the presence of underlying steatohepatitis, severity of disease activity, and fibrosis stage. We further summarize the existing literature around biomarker panels that are derived from interrogation of the metabolome. Their relevance to disease biology and utility in practice are also discussed. We further highlight potential direction for future studies particularly to ensure they are fit for purpose and suitable for widespread use.
Collapse
Affiliation(s)
- En Ying Tan
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore.
| | - Mark D Muthiah
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
4
|
Maclean KN, Jiang H, Neill PD, Chanin RR, Hurt KJ, Orlicky DJ, Bottiglieri T, Roede JR, Stabler SP. Dysregulation of hepatic one-carbon metabolism in classical homocystinuria: Implications of redox-sensitive DHFR repression and tetrahydrofolate depletion for pathogenesis and treatment. FASEB J 2024; 38:e23795. [PMID: 38984928 DOI: 10.1096/fj.202302585r] [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: 12/13/2023] [Revised: 05/30/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024]
Abstract
Cystathionine beta-synthase-deficient homocystinuria (HCU) is a life-threatening disorder of sulfur metabolism. HCU can be treated by using betaine to lower tissue and plasma levels of homocysteine (Hcy). Here, we show that mice with severely elevated Hcy and potentially deficient in the folate species tetrahydrofolate (THF) exhibit a very limited response to betaine indicating that THF plays a critical role in treatment efficacy. Analysis of a mouse model of HCU revealed a 10-fold increase in hepatic levels of 5-methyl -THF and a 30-fold accumulation of formiminoglutamic acid, consistent with a paucity of THF. Neither of these metabolite accumulations were reversed or ameliorated by betaine treatment. Hepatic expression of the THF-generating enzyme dihydrofolate reductase (DHFR) was significantly repressed in HCU mice and expression was not increased by betaine treatment but appears to be sensitive to cellular redox status. Expression of the DHFR reaction partner thymidylate synthase was also repressed and metabolomic analysis detected widespread alteration of hepatic histidine and glutamine metabolism. Many individuals with HCU exhibit endothelial dysfunction. DHFR plays a key role in nitric oxide (NO) generation due to its role in regenerating oxidized tetrahydrobiopterin, and we observed a significant decrease in plasma NOx (NO2 + NO3) levels in HCU mice. Additional impairment of NO generation may also come from the HCU-mediated induction of the 20-hydroxyeicosatetraenoic acid generating cytochrome CYP4A. Collectively, our data shows that HCU induces dysfunctional one-carbon metabolism with the potential to both impair betaine treatment and contribute to multiple aspects of pathogenesis in this disease.
Collapse
Affiliation(s)
- Kenneth N Maclean
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Hua Jiang
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Philip D Neill
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ryan R Chanin
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - K Joseph Hurt
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - David J Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, Texas, USA
| | - James R Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Sally P Stabler
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
5
|
Lambooy S, Heida A, Joschko C, Nakladal D, van Buiten A, Kloosterhuis N, Huijkman N, Gerding A, van de Sluis B, Henning R, Deelman L. Selective Hepatic Cbs Knockout Aggravates Liver Damage, Endothelial Dysfunction and ROS Stress in Mice Fed a Western Diet. Int J Mol Sci 2023; 24:ijms24087019. [PMID: 37108182 PMCID: PMC10138434 DOI: 10.3390/ijms24087019] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/07/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Cystathionine-β-synthase (CBS) is highly expressed in the liver, and deficiencies in Cbs lead to hyperhomocysteinemia (HHCy) and disturbed production of antioxidants such as hydrogen sulfide. We therefore hypothesized that liver-specific Cbs deficient (LiCKO) mice would be particularly susceptible to the development of non-alcoholic fatty liver disease (NAFLD). NAFLD was induced by a high-fat high-cholesterol (HFC) diet; LiCKO and controls were split into eight groups based on genotype (con, LiCKO), diet (normal diet, HFC), and diet duration (12 weeks, 20 weeks). LiCKO mice displayed intermediate to severe HHCy. Plasma H2O2 was increased by HFC, and further aggravated in LiCKO. LiCKO mice fed an HFC diet had heavier livers, increased lipid peroxidation, elevated ALAT, aggravated hepatic steatosis, and inflammation. LiCKO mice showed decreased L-carnitine in the liver, but this did not result in impaired fatty acid oxidation. Moreover, HFC-fed LiCKO mice demonstrated vascular and renal endothelial dysfunction. Liver and endothelial damage correlated significantly with systemic ROS status. In conclusion, this study demonstrates an important role for CBS in the liver in the development of NAFLD, which is most probably mediated through impaired defense against oxidative stress.
Collapse
Affiliation(s)
- Sebastiaan Lambooy
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands
| | - Andries Heida
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands
| | - Christian Joschko
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands
| | - Dalibor Nakladal
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands
| | - Azuwerus van Buiten
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands
| | - Niels Kloosterhuis
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands
| | - Nicolette Huijkman
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands
| | - Albert Gerding
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands
| | - Bart van de Sluis
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands
| | - Robert Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands
| | - Leo Deelman
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9713GZ Groningen, The Netherlands
| |
Collapse
|
6
|
Fernández-Ramos D, Lopitz-Otsoa F, Millet O, Alonso C, Lu SC, Mato JM. One Carbon Metabolism and S-Adenosylmethionine in Non-Alcoholic Fatty Liver Disease Pathogenesis and Subtypes. LIVERS 2022; 2:243-257. [PMID: 37123053 PMCID: PMC10137169 DOI: 10.3390/livers2040020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
One carbon metabolism (1CM) can be defined as the transfer of a carbon unit from one metabolite to another and its replenishment by different sources of labile methyl-group nutrients: primarily choline, methionine, betaine, and serine. This flow of carbon units allows the biosynthesis of nucleotides, amino acids, formylated methionyl-tRNA, polyamines, glutathione, phospholipids, detoxification reactions, maintenance of the redox status and the concentration of NAD, and methylation reactions including epigenetic modifications. That is, 1CM functions as a nutrient sensor and integrator of cellular metabolism. A critical process in 1CM is the synthesis of S-adenosylmethionine (SAMe), the source of essentially all the hundreds of millions of daily methyl transfer reactions in a cell. This versatility of SAMe imposes a tight control in its synthesis and catabolism. Much of our knowledge concerning 1CM has been gained from studies in the production and prevention of nonalcoholic fatty liver disease (NAFLD). Here, we discuss in detail the function of the most important enzymes for their quantitative contribution to maintaining the flux of carbon units through 1CM in the liver and discuss how alterations in their enzymatic activity contribute to the development of NAFLD. Next, we discuss NAFLD subtypes based on serum lipidomic profiles with different risk of cardiovascular disease. Among the latter, we highlight the so-called subtype A for its serum lipidomic profile phenocopying that of mice deficient in SAMe synthesis and because its high frequency (about 50% of the NAFLD patients).
Collapse
Affiliation(s)
- David Fernández-Ramos
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, BRTA, CIBERehd, Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Fernando Lopitz-Otsoa
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, BRTA, CIBERehd, Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, BRTA, CIBERehd, Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Cristina Alonso
- OWL Metabolomics, Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - José M. Mato
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, BRTA, CIBERehd, Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
- Correspondence: ; Tel.: +34-944-061300; Fax: +34-944-0611301
| |
Collapse
|
7
|
Di Minno A, Gelzo M, Caterino M, Costanzo M, Ruoppolo M, Castaldo G. Challenges in Metabolomics-Based Tests, Biomarkers Revealed by Metabolomic Analysis, and the Promise of the Application of Metabolomics in Precision Medicine. Int J Mol Sci 2022; 23:5213. [PMID: 35563604 PMCID: PMC9103094 DOI: 10.3390/ijms23095213] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Abstract
Metabolomics helps identify metabolites to characterize/refine perturbations of biological pathways in living organisms. Pre-analytical, analytical, and post-analytical limitations that have hampered a wide implementation of metabolomics have been addressed. Several potential biomarkers originating from current targeted metabolomics-based approaches have been discovered. Precision medicine argues for algorithms to classify individuals based on susceptibility to disease, and/or by response to specific treatments. It also argues for a prevention-based health system. Because of its ability to explore gene-environment interactions, metabolomics is expected to be critical to personalize diagnosis and treatment. Stringent guidelines have been applied from the very beginning to design studies to acquire the information currently employed in precision medicine and precision prevention approaches. Large, prospective, expensive and time-consuming studies are now mandatory to validate old, and discover new, metabolomics-based biomarkers with high chances of translation into precision medicine. Metabolites from studies on saliva, sweat, breath, semen, feces, amniotic, cerebrospinal, and broncho-alveolar fluid are predicted to be needed to refine information from plasma and serum metabolome. In addition, a multi-omics data analysis system is predicted to be needed for omics-based precision medicine approaches. Omics-based approaches for the progress of precision medicine and prevention are expected to raise ethical issues.
Collapse
Affiliation(s)
- Alessandro Di Minno
- Dipartimento di Farmacia, University of Naples Federico II, 80131 Naples, Italy
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.G.); (M.C.); (M.C.); (M.R.); (G.C.)
| | - Monica Gelzo
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.G.); (M.C.); (M.C.); (M.R.); (G.C.)
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Marianna Caterino
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.G.); (M.C.); (M.C.); (M.R.); (G.C.)
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Michele Costanzo
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.G.); (M.C.); (M.C.); (M.R.); (G.C.)
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Margherita Ruoppolo
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.G.); (M.C.); (M.C.); (M.R.); (G.C.)
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Giuseppe Castaldo
- CEINGE-Biotecnologie Avanzate, 80131 Naples, Italy; (M.G.); (M.C.); (M.C.); (M.R.); (G.C.)
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
8
|
Mateus I, Prip-Buus C. Hydrogen sulphide in liver glucose/lipid metabolism and non-alcoholic fatty liver disease. Eur J Clin Invest 2022; 52:e13680. [PMID: 34519030 PMCID: PMC9285505 DOI: 10.1111/eci.13680] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/24/2021] [Accepted: 09/10/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND For a long time, hydrogen sulphide (H2 S) was considered only as a toxic gas, inhibiting mitochondrial respiration at the level of cytochrome c oxidase, and an environmental pollutant. Nowadays, H2 S is recognized as the third mammalian gasotransmitter, playing an important role in inflammation, septic shock, ischaemia reperfusion events, cardiovascular disease and more recently in liver physiology and chronic liver diseases such as non-alcoholic fatty liver disease (NAFLD). METHODS This narrative review is based on literature search using PubMed. RESULTS From a bioenergetic perspective, H2 S is a very unique molecule, serving as a mitochondrial poison at high concentrations or as an inorganic mitochondrial substrate at low concentrations. By using transgenic animal models to specifically modulate liver H2 S biosynthesis or exogenous compounds that release H2 S, several studies demonstrated that H2 S is a key player in liver glucose and lipid metabolism. Liver H2 S content and biosynthesis were also altered in NAFLD animal models with the in vivo administration of H2 S-releasing molecules preventing the further escalation into non-alcoholic-steatohepatitis. Liver steady-state levels of H2 S, and hence its cell signalling properties, are controlled by a tight balance between its biosynthesis, mainly through the transsulphuration pathway, and its mitochondrial oxidation via the sulphide oxidizing unit. However, studies investigating mitochondrial H2 S oxidation in liver dysfunction still remain scarce. CONCLUSIONS Since H2 S emerges as a key regulator of liver metabolism and metabolic flexibility, further understanding the physiological relevance of mitochondrial H2 S oxidation in liver energy homeostasis and its potential implication in chronic liver diseases are of great interest.
Collapse
Affiliation(s)
- Inês Mateus
- Institut Cochin, INSERM, CNRS, Université de Paris, Paris, France
| | - Carina Prip-Buus
- Institut Cochin, INSERM, CNRS, Université de Paris, Paris, France
| |
Collapse
|
9
|
Gupta S, Wang L, Slifker MJ, Cai KQ, Maclean KN, Wasek B, Bottiglieri T, Kruger WD. Analysis of differential neonatal lethality in cystathionine β-synthase deficient mouse models using metabolic profiling. FASEB J 2021; 35:e21629. [PMID: 33949005 DOI: 10.1096/fj.202100302r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/06/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022]
Abstract
Cystathionine beta-synthase (CBS) is a key enzyme of the trans-sulfuration pathway that converts homocysteine to cystathionine. Loss of CBS activity due to mutation results in CBS deficiency, an inborn error of metabolism characterized by extreme elevation of plasma total homocysteine (tHcy). C57BL6 mice containing either a homozygous null mutation in the cystathionine β-synthase (Cbs-/- ) gene or an inactive human CBS protein (Tg-G307S Cbs-/- ) are born in mendelian numbers, but the vast majority die between 18 and 21 days of age due to liver failure. However, adult Cbs null mice that express a hypomorphic allele of human CBS as a transgene (Tg-I278T Cbs-/- ) show almost no neonatal lethality despite having serum tHcy levels similar to mice with no CBS activity. Here, we characterize liver and serum metabolites in neonatal Cbs+/- , Tg-G307S Cbs-/- , and Tg-I278T Cbs-/- mice at 6, 10, and 17 days of age to understand this difference. In serum, we observe similar elevations in tHcy in both Tg-G307S Cbs-/- and Tg-I278T Cbs-/- compared to control animals, but methionine is much more severely elevated in Tg-G307S Cbs-/- mice. Large scale metabolomic analysis of liver tissue confirms that both methionine and methionine-sulfoxide are significantly more elevated in Tg-G307S Cbs-/- animals, along with significant differences in several other metabolites including hexoses, amino acids, other amines, lipids, and carboxylic acids. Our data are consistent with a model that the neonatal lethality observed in CBS-null mice is driven by excess methionine resulting in increased stress on a variety of related pathways including the urea cycle, TCA cycle, gluconeogenesis, and phosphatidylcholine biosynthesis.
Collapse
Affiliation(s)
- Sapna Gupta
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Liqun Wang
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Michael J Slifker
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Kathy Q Cai
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Kenneth N Maclean
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Brandi Wasek
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX, USA
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX, USA
| | - Warren D Kruger
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
10
|
Maclean KN, Jiang H, Phinney WN, Mclagan BM, Roede JR, Stabler SP. Derangement of hepatic polyamine, folate, and methionine cycle metabolism in cystathionine beta-synthase-deficient homocystinuria in the presence and absence of treatment: Possible implications for pathogenesis. Mol Genet Metab 2021; 132:128-138. [PMID: 33483253 DOI: 10.1016/j.ymgme.2021.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 11/22/2022]
Abstract
Cystathionine beta-synthase deficient homocystinuria (HCU) is a life-threatening disorder of sulfur metabolism. Our knowledge of the metabolic changes induced in HCU are based almost exclusively on data derived from plasma. In the present study, we present a comprehensive analysis on the effects of HCU upon the hepatic metabolites and enzyme expression levels of the methionine-folate cycles in a mouse model of HCU. HCU induced a 10-fold increase in hepatic total homocysteine and in contrast to plasma, this metabolite was only lowered by approximately 20% by betaine treatment indicating that this toxic metabolite remains unacceptably elevated. Hepatic methionine, S-adenosylmethionine, S-adenosylhomocysteine, N-acetlymethionine, N-formylmethionine, methionine sulfoxide, S-methylcysteine, serine, N-acetylserine, taurocyamine and N-acetyltaurine levels were also significantly increased by HCU while cysteine, N-acetylcysteine and hypotaurine were all significantly decreased. In terms of polyamine metabolism, HCU significantly decreased spermine and spermidine levels while increasing 5'-methylthioadenosine. Betaine treatment restored normal spermine and spermidine levels but further increased 5'-methylthioadenosine. HCU induced a 2-fold induction in expression of both S-adenosylhomocysteine hydrolase and methylenetetrahydrofolate reductase. Induction of this latter enzyme was accompanied by a 10-fold accumulation of its product, 5-methyl-tetrahydrofolate, with the potential to significantly perturb one‑carbon metabolism. Expression of the cytoplasmic isoform of serine hydroxymethyltransferase was unaffected by HCU but the mitochondrial isoform was repressed indicating differential regulation of one‑carbon metabolism in different sub-cellular compartments. All HCU-induced changes in enzyme expression were completely reversed by either betaine or taurine treatment. Collectively, our data show significant alterations of polyamine, folate and methionine cycle metabolism in HCU hepatic tissues that in some cases, differ significantly from those observed in plasma, and have the potential to contribute to multiple aspects of pathogenesis.
Collapse
Affiliation(s)
- Kenneth N Maclean
- Departments of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Hua Jiang
- Departments of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Whitney N Phinney
- Medicine and University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Bailey M Mclagan
- Departments of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - James R Roede
- Pharmaceutical Sciences, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Sally P Stabler
- Medicine and University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
11
|
Xu J, Huang X. Lipid Metabolism at Membrane Contacts: Dynamics and Functions Beyond Lipid Homeostasis. Front Cell Dev Biol 2020; 8:615856. [PMID: 33425923 PMCID: PMC7786193 DOI: 10.3389/fcell.2020.615856] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 11/30/2020] [Indexed: 01/12/2023] Open
Abstract
Membrane contact sites (MCSs), regions where the membranes of two organelles are closely apposed, play critical roles in inter-organelle communication, such as lipid trafficking, intracellular signaling, and organelle biogenesis and division. First identified as “fraction X” in the early 90s, MCSs are now widely recognized to facilitate local lipid synthesis and inter-organelle lipid transfer, which are important for maintaining cellular lipid homeostasis. In this review, we discuss lipid metabolism and related cellular and physiological functions in MCSs. We start with the characteristics of lipid synthesis and breakdown at MCSs. Then we focus on proteins involved in lipid synthesis and turnover at these sites. Lastly, we summarize the cellular function of lipid metabolism at MCSs beyond mere lipid homeostasis, including the physiological meaning and relevance of MCSs regarding systemic lipid metabolism. This article is part of an article collection entitled: Coupling and Uncoupling: Dynamic Control of Membrane Contacts.
Collapse
Affiliation(s)
- Jiesi Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
12
|
Di Minno A, Anesi A, Chiesa M, Cirillo F, Colombo GI, Orsini RC, Capasso F, Morisco F, Fiorelli S, Eligini S, Cavalca V, Tremoli E, Porro B, Di Minno MND. Plasma phospholipid dysregulation in patients with cystathionine-β synthase deficiency. Nutr Metab Cardiovasc Dis 2020; 30:2286-2295. [PMID: 32912785 DOI: 10.1016/j.numecd.2020.07.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 01/27/2023]
Abstract
BACKGROUND & AIMS Patients with cystathionine β-synthase deficiency (CBSD) exhibit high circulating levels of homocysteine and enhanced lipid peroxidation. We have characterized the plasma lipidome in CBSD patients and related lipid abnormalities with reactions underlying enhanced homocysteine levels. METHODS AND RESULTS Using an ultra-high-performance liquid chromatography-electrospray ionization-quadrupole-time of flight-mass spectrometry method, plasma lipids were determined with an untargeted lipidomics approach in 11 CBSD patients and 11 matched healthy subjects (CTRL). Compared to CTRL, CBSD patients had a higher medium and long-chain polyunsaturated fatty acids (PUFA) content in phosphatidylethanolamine (PE) and lysophosphatidylethanolamine (LPE) species (p < 0.02), and depletion of phosphatidylcholine (PC; p = 0.02) and of lysophosphatidylcholine (LPC; p = 0.003) species containing docosahexaenoic acid (DHA), suggesting impaired phosphatidylethanolamine-N-methyltransferase (PEMT) activity. PEMT converts PE into PC using methyl group by S-adenosylmethionine (SAM) thus converted in S-adenosylhomocysteine (SAH). Whole blood SAM and SAH concentrations by liquid chromatography tandem mass spectrometry were 1.4-fold (p = 0.015) and 5.3-fold (p = 0.003) higher in CBSD patients than in CTRL. A positive correlation between SAM/SAH and PC/PE ratios (r = 0.520; p = 0.019) was found. CONCLUSIONS A novel biochemical abnormality in CBSD patients consisting in depletion of PC and LPC species containing DHA and accumulation of PUFA in PE and LPE species is revealed by this lipidomic approach. Changes in plasma SAM and SAH concentrations are associated with such phospholipid dysregulation. Given the key role of DHA in thrombosis prevention, depletion of PC species containing DHA in CBSD patients provides a new direction to understand the poor cardiovascular outcome of patients with homocystinuria.
Collapse
Affiliation(s)
- Alessandro Di Minno
- Dipartimento di Farmacia, Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Andrea Anesi
- Fondazione Edmund Mach Research and Innovation Centre, Food Quality and Nutrition Department, S. Michele all' Adige, Trento, Italy
| | | | - Ferdinando Cirillo
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy
| | | | - Roberta C Orsini
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy
| | - Filomena Capasso
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy
| | - Filomena Morisco
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy
| | | | | | | | | | | | - Matteo N D Di Minno
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy
| |
Collapse
|
13
|
da Silva RP, Eudy BJ, Deminice R. One-Carbon Metabolism in Fatty Liver Disease and Fibrosis: One-Carbon to Rule Them All. J Nutr 2020; 150:994-1003. [PMID: 32119738 DOI: 10.1093/jn/nxaa032] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/14/2019] [Accepted: 01/30/2020] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a term used to characterize a range of disease states that involve the accumulation of fat in the liver but are not associated with excessive alcohol consumption. NAFLD is a prevalent disease that can progress to organ damage like liver cirrhosis and hepatocellular carcinoma. Many animal models have demonstrated that one-carbon metabolism is strongly associated with NAFLD. Phosphatidylcholine is an important phospholipid that affects hepatic lipid homeostasis and de novo synthesis of this phospholipid is associated with NAFLD. However, one-carbon metabolism serves to support all cellular methylation reactions and catabolism of methionine, serine, glycine, choline, betaine, tryptophan, and histidine. Several different pathways within one-carbon metabolism that play important roles in regulating energy metabolism and immune function have received less attention in the study of fatty liver disease and fibrosis. This review examines what we have learned about hepatic lipid metabolism and liver damage from the study of one-carbon metabolism thus far and highlights unexplored opportunities for future research.
Collapse
Affiliation(s)
- Robin P da Silva
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, USA
| | - Brandon J Eudy
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, USA
| | - Rafael Deminice
- Department of Physical Education, State University of Londrina, Londrina, Paraná, Brazil
| |
Collapse
|
14
|
Wang J, He W, Tsai PJ, Chen PH, Ye M, Guo J, Su Z. Mutual interaction between endoplasmic reticulum and mitochondria in nonalcoholic fatty liver disease. Lipids Health Dis 2020; 19:72. [PMID: 32284046 PMCID: PMC7155254 DOI: 10.1186/s12944-020-01210-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common metabolic syndrome. Imbalances between liver lipid output and input are the direct causes of NAFLD, and hepatic steatosis is the pathological premise and basis for NAFLD progression. Mutual interaction between endoplasmic reticulum stress (ERS) and oxidative stress play important roles in NAFLD pathogenesis. Notably, mitochondria-associated membranes (MAMs) act as a structural bridges for functional clustering of molecules, particularly for Ca2+, lipids, and reactive oxygen species (ROS) exchange. Previous studies have examined the crucial roles of ERS and ROS in NAFLD and have shown that MAM structural and functional integrity determines normal ER- mitochondria communication. Upon disruption of MAM integrity, miscommunication directly or indirectly causes imbalances in Ca2+ homeostasis and increases ERS and oxidative stress. Here, we emphasize the involvement of MAMs in glucose and lipid metabolism, chronic inflammation and insulin resistance in NAFLD and summarize MAM-targeting drugs and compounds, most of which achieve their therapeutic or ameliorative effects on NAFLD by improving MAM integrity. Therefore, targeting MAMs may be a viable strategy for NAFLD treatment. This review provides new ideas and key points for basic NAFLD research and drug development centred on mitochondria and the endoplasmic reticulum.
Collapse
Affiliation(s)
- Jin Wang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Key Laboratory of Modulating Liver to Treat Hyperlipemia SATCM, Level 3 Laboratory of Lipid Metabolism SATCM, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Wanping He
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Key Laboratory of Modulating Liver to Treat Hyperlipemia SATCM, Level 3 Laboratory of Lipid Metabolism SATCM, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ping-Ju Tsai
- King-Prebiotics Biotechnology (TW) Co., LTD, 2F.-1, No. 250, Zhongshan Rd., Linkou Dist, New Taipei City, 24446, Taiwan
| | - Pei-Hsuan Chen
- King-Prebiotics Biotechnology (TW) Co., LTD, 2F.-1, No. 250, Zhongshan Rd., Linkou Dist, New Taipei City, 24446, Taiwan
| | - Manxiang Ye
- New Francisco (Yunfu City) Biotechnology Co, Ltd Swan-kan-chiau Ind. Dist., Kaofong Village, Yunfu City, Guangdong, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Key Laboratory of Modulating Liver to Treat Hyperlipemia SATCM, Level 3 Laboratory of Lipid Metabolism SATCM, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
15
|
George AK, Homme RP, Majumder A, Laha A, Metreveli N, Sandhu HS, Tyagi SC, Singh M. Hydrogen sulfide intervention in cystathionine-β-synthase mutant mouse helps restore ocular homeostasis. Int J Ophthalmol 2019; 12:754-764. [PMID: 31131233 DOI: 10.18240/ijo.2019.05.09] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 02/24/2019] [Indexed: 02/03/2023] Open
Abstract
AIM To investigate the applications of hydrogen sulfide (H2S) in eye-specific ailments in mice. METHODS Heterozygous cystathionine-β-synthase (CBS+/-) and wild-type C57BL/6J (WT) mice fed with or without high methionine diet (HMD) were administered either phosphate buffered saline (PBS) or the slow-release H2S donor: GYY4137. Several analyses were performed to study GYY4137 effects by examining retinal lysates for key protein expressions along with plasma glutamate and glutathione estimations. Intraocular pressure (IOP) was monitored during GYY4137 treatment; barium sulfate and bovine serum albumin conjugated fluorescein isothiocyanate (BSA-FITC) angiographies were performed for examining vasculature and its permeability post-treatment. Vision-guided behavior was also tested employing novel object recognition test (NORT) and light-dark box test (LDBT) recordings. RESULTS CBS deficiency (CBS+/-) coupled with HMD led disruption of methionine/homocysteine (Hcy) metabolism leading to hyperhomocysteinemia (HHcy) in CBS+/- mice as reflected by increased Hcy, and s-adenosylhomocysteine hydrolase (SAHH) levels. Unlike CBS, cystathionine-γ lyase (CSE), methylenetetrahydrofolate reductase (MTHFR) levels which were reduced but compensated by GYY4137 intervention. Heightened oxidative and endoplasmic reticulum (ER) stress responses were mitigated by GYY4137 effects along with enhanced glutathione (GSH) levels. Increased glutamate levels in CBS+/- strain were prominent than WT mice and these mice also exhibited higher IOP that was lowered by GYY4137 treatment. CBS deficiency also resulted in vision-guided behavioral impairment as revealed by NORT and LDBT findings. Interestingly, GYY4137 was able to improve CBS+/- mice behavior together with lowering their glutamate levels. Blood-retinal barrier (BRB) appeared compromised in CBS+/- with vessels' leakage that was mitigated in GYY4137 treated group. This corroborated the results for occludin (an integral plasma membrane protein of the cellular tight junctions) stabilization. CONCLUSION Findings reveal that HHcy-induced glutamate excitotoxicity, oxidative damage, ER-stress and vascular permeability alone or together can compromise ocular health and that GYY4137 could serve as a potential therapeutic agent for treating HHcy induced ocular disorders.
Collapse
Affiliation(s)
- Akash K George
- Eye and Vision Science Laboratory, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.,Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | - Rubens P Homme
- Eye and Vision Science Laboratory, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.,Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | - Avisek Majumder
- Department of Medicine, UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA
| | - Anwesha Laha
- Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | - Naira Metreveli
- Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | - Harpal S Sandhu
- Department of Ophthalmology and Visual Sciences; Kentucky Lions Eye Center, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | - Mahavir Singh
- Eye and Vision Science Laboratory, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.,Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| |
Collapse
|
16
|
Yang A, Jiao Y, Yang S, Deng M, Yang X, Mao C, Sun Y, Ding N, Li N, Zhang M, Jin S, Zhang H, Jiang Y. Homocysteine activates autophagy by inhibition of CFTR expression via interaction between DNA methylation and H3K27me3 in mouse liver. Cell Death Dis 2018; 9:169. [PMID: 29415998 PMCID: PMC5833451 DOI: 10.1038/s41419-017-0216-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/11/2017] [Accepted: 12/13/2017] [Indexed: 12/12/2022]
Abstract
Elevated homocysteine (Hcy) levels have been reported to be involved in liver injury, and autophagy plays an important role in normal hepatic physiology and pathophysiology, but the mechanism underlying Hcy regulated autophagy is currently unknown. In this study, CBS+/- mice were fed with regular diet for 12 weeks to establish a hyperhomocysteinemia (HHcy) model and HL-7702 cells were treated with Hcy, we found that Hcy increases autophagy and aggravates liver injury by downregulation of cystic fibrosis transmembrane conductance regulator (CFTR) expression in vivo and in vitro. Overexpression of CFTR inhibited the formation of autophagosomes and the expression of autophagy-related proteins BECN1, LC3-II/I and Atg12, while the expression of p62 increased in Hcy-treated hepatocytes and CBS+/- mice injected with lentivirus expressing CFTR. Further study showed that CFTR expression is regulated by the interaction of DNA methyltransferase 1 (DNMT1) and enhancer of zeste homolog 2 (EZH2), which, respectively, regulate DNA methylation and histone H3 lysine 27 trimethylation (H3K27me3). In conclusion, our study showed that Hcy activates autophagy by inhibition of CFTR expression via interaction between H3K27me3 and DNA methylation in the mouse liver. These findings provide new insight into the mechanism of Hcy-induced autophagy in liver injury.
Collapse
Affiliation(s)
- Anning Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.,Ningxia Key Laboratory of Vascular Injury and Repair Research, Yinchuan, China
| | - Yun Jiao
- Ningxia Medical University General Hospital, Yinchuan, 750004, China
| | - Songhao Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.,Ningxia Key Laboratory of Vascular Injury and Repair Research, Yinchuan, China
| | - Mei Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.,Ningxia Key Laboratory of Vascular Injury and Repair Research, Yinchuan, China
| | - Xiaoling Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.,Ningxia Key Laboratory of Vascular Injury and Repair Research, Yinchuan, China
| | - Caiyan Mao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.,Ningxia Key Laboratory of Vascular Injury and Repair Research, Yinchuan, China
| | - Yue Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.,Ningxia Key Laboratory of Vascular Injury and Repair Research, Yinchuan, China
| | - Ning Ding
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.,Ningxia Key Laboratory of Vascular Injury and Repair Research, Yinchuan, China
| | - Nan Li
- Pharmacy college, Ningxia Medical University, Yinchuan, 750004, China
| | - Minghao Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.,Ningxia Key Laboratory of Vascular Injury and Repair Research, Yinchuan, China
| | - Shaoju Jin
- Pharmacy college, Ningxia Medical University, Yinchuan, 750004, China
| | - Huiping Zhang
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Yinchuan, China. .,Ningxia Medical University General Hospital, Yinchuan, 750004, China.
| | - Yideng Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China. .,Ningxia Key Laboratory of Vascular Injury and Repair Research, Yinchuan, China.
| |
Collapse
|
17
|
Kruger WD. Cystathionine β-synthase deficiency: Of mice and men. Mol Genet Metab 2017; 121:199-205. [PMID: 28583326 PMCID: PMC5526210 DOI: 10.1016/j.ymgme.2017.05.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 01/28/2023]
Abstract
Cystathionine β-synthase (CBS) deficiency (Online Mendelian Inheritance in Man [OMIM] 236,200) is an autosomal recessive disorder that is caused by mutations in the CBS gene. It is the most common inborn error of sulfur metabolism and is the cause of classical homocystinuria, a condition characterized by very high levels of plasma total homocysteine and methionine. Although recognized as an inborn error of metabolism over 60years ago, these is still much we do not understand related to how this specific metabolic defect gives rise to its distinct phenotypes. To try and answer these questions, several groups have developed mouse models on CBS deficiency. In this article, we will review various mouse models of CBS deficiency and discuss how these mouse models compare to human CBS deficient patients.
Collapse
Affiliation(s)
- Warren D Kruger
- Cancer Biology Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA.
| |
Collapse
|