1
|
Savulescu-Fiedler I, Dorobantu-Lungu LR, Dragosloveanu S, Benea SN, Dragosloveanu CDM, Caruntu A, Scheau AE, Caruntu C, Scheau C. The Cross-Talk Between the Peripheral and Brain Cholesterol Metabolisms. Curr Issues Mol Biol 2025; 47:115. [PMID: 39996836 PMCID: PMC11853762 DOI: 10.3390/cimb47020115] [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: 12/21/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Cholesterol is an essential element for the development and normal function of the central nervous system. While peripheral cholesterol is influenced by liver metabolism and diet, brain cholesterol metabolism takes place in an isolated system due to the impermeability of the blood-brain barrier (BBB). However, cross-talk occurs between the brain and periphery, specifically through metabolites such as oxysterols that play key roles in regulating cholesterol balance. Several neurodegenerative conditions such as Alzheimer's disease or Parkinson's disease are considered to be affected by the loss of this balance. Also, the treatment of hypercholesterolemia needs to consider these discrete interferences between brain and peripheral cholesterol and the possible implications of each therapeutic approach. This is particularly important because of 27-hydroxycholesterol and 24-hydroxycholesterol, which can cross the BBB and are involved in cholesterol metabolism. This paper examines the metabolic pathways of cholesterol metabolism in the brain and periphery and focuses on the complex cross-talk between these metabolisms. Also, we emphasize the regulatory role of the BBB and the need for an integrated approach to cholesterol management.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Luiza-Roxana Dorobantu-Lungu
- Department of Cardiology, Emergency Institute for Cardiovascular Diseases “C.C. Iliescu”, 022328 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Departament of Infectious Diseases, National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, 021105 Bucharest, Romania
| | - Christiana Diana Maria Dragosloveanu
- Department of Ophthalmology, Faculty of Dentistry, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Ophthalmology, Clinical Hospital for Ophthalmological Emergencies, 010464 Bucharest, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| |
Collapse
|
2
|
Yan G, Qin Z, Liu A, Huang Z, Wang X, Zhang S, Xie X, Huang X, Chen J, Li Y, Xie Q, Liu Y, Su Z, Xie J. Sulfonation metabolism in the gut microbiota is the main metabolic pathway of cholesterol in hypercholesterolemic mice. Food Funct 2024; 15:9750-9765. [PMID: 39238326 DOI: 10.1039/d4fo02312a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
The interactions between dietary cholesterol and intestinal microbiota strongly affect host health. Sulfonation is a major conjugating pathway responsible for regulating the chemical and functional homeostasis of endogenous and exogenous molecules. However, the role of cholesterol sulfonation metabolism in the host remains unclear. This work was designed to profile cholesterol-specific host-microbe interaction and conversion focusing on cholesterol sulfonation metabolism. Results indicated that the serum and fecal cholesterol sulfate (CHS) levels were significantly higher than those of total bile acid (TBA) levels in hypercholesterolemic mice. Deletion of the gut microbiota by antibiotics could dramatically increase total cholesterol (TC) levels but it decreased CHS levels in a pseudo-germ-free (PGF) mouse host. 16S rRNA gene sequencing assay and correlation analysis between the abundance of various intestinal bacteria (phylum and class) and the CHS/TC ratio showed that the intestinal genera Bacteroides contributed essentially to cholesterol sulfonation metabolism. These results were further confirmed in an in situ and ex vivo mouse intestinal model, which indicated that the sulfonation metabolism rate of cholesterol could reach 42% under high cholesterol conditions. These findings provided new evidence that the sulfonation metabolic pathway dominated cholesterol metabolism in hypercholesterolemic mice and microbial conversion of cholesterol-to-CHS was of vital importance for cholesterol-lowering by Bacteroides. This suggested that the gut microbiota could regulate cholesterol metabolism and that it was feasible to reduce cholesterol levels by dietary interventions involving the gut microbiota.
Collapse
Affiliation(s)
- Guangtao Yan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Zehui Qin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, PR China
| | - Aitong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Ziwei Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Xinhong Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Shanli Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Xiaolin Xie
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Xiaoqi Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Qingfeng Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China.
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Jianhui Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China.
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120, PR China
| |
Collapse
|
3
|
Alherz FA. Human sulfotransferase SULT2B1 physiological role and the impact of genetic polymorphism on enzyme activity and pathological conditions. Front Genet 2024; 15:1464243. [PMID: 39280099 PMCID: PMC11392796 DOI: 10.3389/fgene.2024.1464243] [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: 07/13/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024] Open
Abstract
Human SULT2B1gene is responsible for expressing SULT2B1a and SULT2B1b enzymes, which are phase II metabolizing enzymes known as pregnenolone and cholesterol sulfotransferase (SULT), respectively. They are expressed in several tissues and contribute to steroids and hydroxysteroids homeostasis. Genetic variation of the SULT2B1 is reported to be associated with various pathological conditions, including autosomal recessive ichthyosis, cardiovascular disease, and different types of cancers. Understanding the pathological impact of SULT2B1 genetic polymorphisms in the human body is crucial to incorporating these findings in evaluating clinical conditions or improving therapeutic efficacy. Therefore, this paper summarized the most relevant reported studies concerning SULT2B1 expression, tissue distribution, substrates, and reported genetic polymorphisms and their mechanisms in enzyme activity and pathological conditions.
Collapse
Affiliation(s)
- Fatemah A Alherz
- Department of Pharmaceutical Science, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| |
Collapse
|
4
|
D'Agostino GD, Chaudhari SN, Devlin AS. Host-microbiome orchestration of the sulfated metabolome. Nat Chem Biol 2024; 20:410-421. [PMID: 38347214 PMCID: PMC11095384 DOI: 10.1038/s41589-023-01526-9] [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: 12/23/2022] [Accepted: 12/08/2023] [Indexed: 04/01/2024]
Abstract
Recent studies have demonstrated that metabolites produced by commensal bacteria causally influence health and disease. The sulfated metabolome is one class of molecules that has recently come to the forefront due to efforts to understand the role of these metabolites in host-microbiome interactions. Sulfated compounds have canonically been classified as waste products; however, studies have revealed a variety of physiological roles for these metabolites, including effects on host metabolism, immune response and neurological function. Moreover, recent research has revealed that commensal bacteria either chemically modify or synthesize a variety of sulfated compounds. In this Review, we explore how host-microbiome collaborative metabolism transforms the sulfated metabolome. We describe bacterial and mammalian enzymes that sulfonate and desulfate biologically relevant carbohydrates, amino acid derivatives and cholesterol-derived metabolites. We then discuss outstanding questions and future directions in the field, including potential roles of sulfated metabolites in disease detection, prevention and treatment. We hope that this Review inspires future research into sulfated compounds and their effects on physiology.
Collapse
Affiliation(s)
- Gabriel D D'Agostino
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Snehal N Chaudhari
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
- Department of Biochemistry, University of Wisconsin, Madison, WI, USA.
| | - A Sloan Devlin
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Duffel MW, Lehmler HJ. Complex roles for sulfation in the toxicities of polychlorinated biphenyls. Crit Rev Toxicol 2024; 54:92-122. [PMID: 38363552 PMCID: PMC11067068 DOI: 10.1080/10408444.2024.2311270] [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: 12/29/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/17/2024]
Abstract
Polychlorinated biphenyls (PCBs) are persistent organic toxicants derived from legacy pollution sources and their formation as inadvertent byproducts of some current manufacturing processes. Metabolism of PCBs is often a critical component in their toxicity, and relevant metabolic pathways usually include their initial oxidation to form hydroxylated polychlorinated biphenyls (OH-PCBs). Subsequent sulfation of OH-PCBs was originally thought to be primarily a means of detoxication; however, there is strong evidence that it may also contribute to toxicities associated with PCBs and OH-PCBs. These contributions include either the direct interaction of PCB sulfates with receptors or their serving as a localized precursor for OH-PCBs. The formation of PCB sulfates is catalyzed by cytosolic sulfotransferases, and, when transported into the serum, these metabolites may be retained, taken up by other tissues, and subjected to hydrolysis catalyzed by intracellular sulfatase(s) to regenerate OH-PCBs. Dynamic cycling between PCB sulfates and OH-PCBs may lead to further metabolic activation of the resulting OH-PCBs. Ultimate toxic endpoints of such processes may include endocrine disruption, neurotoxicities, and many others that are associated with exposures to PCBs and OH-PCBs. This review highlights the current understanding of the complex roles that PCB sulfates can have in the toxicities of PCBs and OH-PCBs and research on the varied mechanisms that control these roles.
Collapse
Affiliation(s)
- Michael W. Duffel
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, Iowa, 52242, United States
| | - Hans-Joachim Lehmler
- Department of Occupational and Environmental Health, College of Public Health, The University of Iowa, Iowa City, Iowa, 52242, United States
| |
Collapse
|
6
|
Park JS, Chung IJ, Kim HR, Jun CD. The Immunosuppressive Potential of Cholesterol Sulfate Through T Cell Microvilli Disruption. Immune Netw 2023; 23:e29. [PMID: 37416932 PMCID: PMC10320417 DOI: 10.4110/in.2023.23.e29] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/28/2023] [Accepted: 04/21/2023] [Indexed: 07/08/2023] Open
Abstract
Cholesterol (CL) is required for various biomolecular production processes, including those of cell membrane components. Therefore, to meet these needs, CL is converted into various derivatives. Among these derivatives is cholesterol sulfate (CS), a naturally produced CL derivative by the sulfotransferase family 2B1 (SULT2B1), which is widely present in human plasma. CS is involved in cell membrane stabilization, blood clotting, keratinocyte differentiation, and TCR nanocluster deformation. This study shows that treatment of T cells with CS resulted in the decreased surface expression of some surface T-cell proteins and reduced IL-2 release. Furthermore, T cells treated with CS significantly reduced lipid raft contents and membrane CLs. Surprisingly, using the electron microscope, we also observed that CS led to the disruption of T-cell microvilli, releasing small microvilli particles containing TCRs and other microvillar proteins. However, in vivo, T cells with CS showed aberrant migration to high endothelial venules and limited infiltrating splenic T-cell zones compared with the untreated T cells. Additionally, we observed significant alleviation of atopic dermatitis in mice injected with CS in the animal model. Based on these results, we conclude that CS is an immunosuppressive natural lipid that impairs TCR signaling by disrupting microvillar function in T cells, suggesting its usefulness as a therapeutic agent for alleviating T-cell-mediated hypersensitivity and a potential target for treating autoimmune diseases.
Collapse
Affiliation(s)
- Jeong-Su Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Ik-Joo Chung
- Department of Hematology-Oncology, Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Hye-Ran Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Division of Rare and Refractory Cancer, Tumor Immunology, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| |
Collapse
|
7
|
Wang SS, Xu X, Lu AX, Li WH, Liu JX, Liu C, Yan CH. Amelioration of cholesterol sulfate for lead-induced CTX cell apoptosis based on BDNF signaling pathway mediated cholesterol metabolism. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 248:114307. [PMID: 36423370 DOI: 10.1016/j.ecoenv.2022.114307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 06/16/2023]
Abstract
Lead (Pb), as a deleterious heavy metal, ubiquitously exists in environment and industry, which engenders multi-organ disfunction, especially the brain of infants who are vulnerable to attack from lead-induced neurotoxicity. Although cholesterol sulfate (CS) is crucial constituent of cell membranes and precursor of neurosteroids, which maintains the function and survival of neurons, the role of CS in lead-induced neurological damage still remains incomplete. In this work, Rat Brain Astrocytes cell line (CTX cells) was applied into exploration that protective effects of CS on CTX cell apoptosis induced by lead via the regulation of BDNF/TrkB signaling pathway mediated cholesterol metabolism. We found that CTX cells exposed to lead manifested apparent cytotoxicity, where the viability of CTX cells was significantly suppressed, accompanied with the elevation of apoptosis, in response to a trend towards increases in reactive oxygen species (ROS) production and pro-apoptotic protein Cleaved-caspase3, synchronized with the decline in anti-apoptotic protein Bcl-2. Moreover, accumulation of lead in CTX cells showed a dose-dependent increase, and meanwhile, decrements in cholesterol content occurred along with increase in lead exposure, in which expressions of cholesterol metabolism related proteins and transcriptions of its genes (SREBP2, LDLR, and HMGCR) were diminished. Furthermore, BDNF signaling pathway was obviously blocked after lead exposure, down-regulating expressions of proteins BDNF and TrkB. However, pretreatment with CS detoxified the negative impacts of lead-invoked CTX cell damage, acting as an effective remedy for apoptosis, imbalance of cholesterol metabolism and inhibition of BDNF signaling pathway. In addition, the relationship between BDNF signaling pathway and cholesterol metabolism was further verified, in which cholesterol metabolism related proteins and genes were promoted significantly after the activation of BDNF/TrkB signaling pathway using 7,8-Dihydroxyflavone (7,8-DHF), thereby detoxifying lead-induced CTX cell injury. However, the pretreatment of TrkB inhibitor ANA-12 offset the promotion of 7,8-DHF and ultimately inhibit cholesterol metabolism. Overall, our study demonstrated that CS could initiate the BDNF/TrkB signaling pathway, regulating the cholesterol metabolism against CTX cell apoptosis invoked by lead.
Collapse
Affiliation(s)
- Su-Su Wang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; School of Public Health, Shanghai Jiao Tong University, Shanghai, China
| | - Xi Xu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - An-Xin Lu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wan-He Li
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jun-Xia Liu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Liu
- School of Public Health, Xuzhou Medical University, Jiangsu, China
| | - Chong-Huai Yan
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
8
|
Abdalkareem Jasim S, Kzar HH, Haider Hamad M, Ahmad I, Al-Gazally ME, Ziyadullaev S, Sivaraman R, Abed Jawad M, Thaeer Hammid A, Oudaha KH, Karampoor S, Mirzaei R. The emerging role of 27-hydroxycholesterol in cancer development and progression: An update. Int Immunopharmacol 2022; 110:109074. [PMID: 35978522 DOI: 10.1016/j.intimp.2022.109074] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/09/2022] [Accepted: 07/17/2022] [Indexed: 02/07/2023]
Abstract
Oxysterols are cholesterol metabolites generated in the liver and other peripheral tissues as a mechanism of removing excess cholesterol. Oxysterols have a wide range of biological functions, including the regulation of sphingolipid metabolism, platelet aggregation, and apoptosis. However, it has been found that metabolites derived from cholesterol play essential functions in cancer development and immunological suppression. In this regard, research indicates that 27-hydroxycholesterol (27-HC) might act as an estrogen, promoting the growth of estrogen receptor (ER) positive breast cancer cells. The capacity of cholesterol to dynamically modulate signaling molecules inside the membrane and particular metabolites serving as signaling molecules are two possible contributory processes. 27-HC is a significant metabolite produced mainly through the CYP27A1 (Cytochrome P450 27A1) enzyme. 27-HC maintains cholesterol balance biologically by promoting cholesterol efflux via the liver X receptor (LXR) and suppressing de novo cholesterol production through the Insulin-induced Genes (INSIGs). It has been demonstrated that 27-HC is able to function as a selective ER regulator. Moreover, enhanced 27-HC production is in favor of the growth of end-stage malignancies in the brain, thyroid organs, and colon, as shown in breast cancer, probably due to pro-survival and pro-inflammatory signaling induced by unbalanced levels of oxysterols. However, the actual role of 27-HC in cancer promotion and progression remains debatable, and many studies are warranted to be performed to unravel the precise function of these molecules. This review article will summarize the latest evidence on the deleterious or beneficial functions of 27-HC in various types of cancer, such as breast cancer, prostate cancer, colon cancer, gastric cancer, ovarian cancer, endometrial cancer, lung cancer, melanoma, glioblastoma, thyroid cancer, adrenocortical cancer, and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-maarif University College, Al-anbar-Ramadi, Iraq
| | - Hamzah H Kzar
- Veterinary medicine college, Al-Qasim green University, Al-Qasim, Iraq
| | - Mohammed Haider Hamad
- Medical Laboratory Techniques Department, Al Mustaqbal University college, Babylon, Iraq
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Shukhrat Ziyadullaev
- Professor, Doctor of Medical Sciences, No.1 Department of Internal Diseases, Vice-rector for Scientific Affairs and Innovations, Samarkand State Medical University, Amir Temur Street 18, Samarkand, Uzbekistan
| | - R Sivaraman
- Department of Mathematics, Institution of Dwaraka Doss Goverdhan Doss Vaishnav College, Arumbakkam, Chennai, University of Madras, Chennai, India
| | | | - Ali Thaeer Hammid
- Computer Engineering Techniques Department, Faculty of Information Technology, Imam Ja'afar Al-Sadiq University, Baghdad, Iraq
| | - Khulood H Oudaha
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University Thi-Qar, Iraq
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
9
|
Massey WJ, Brown JM. Microbial regulation of cholesterol homeostasis. Nat Microbiol 2022; 7:1327-1328. [PMID: 35982309 DOI: 10.1038/s41564-022-01186-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- William J Massey
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH, USA. .,Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH, USA.
| | - J Mark Brown
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH, USA. .,Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
10
|
Yao L, D'Agostino GD, Park J, Hang S, Adhikari AA, Zhang Y, Li W, Avila-Pacheco J, Bae S, Clish CB, Franzosa EA, Huttenhower C, Huh JR, Devlin AS. A biosynthetic pathway for the selective sulfonation of steroidal metabolites by human gut bacteria. Nat Microbiol 2022; 7:1404-1418. [PMID: 35982310 DOI: 10.1038/s41564-022-01176-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 06/20/2022] [Indexed: 12/13/2022]
Abstract
Members of the human gut microbiome enzymatically process many bioactive molecules in the gastrointestinal tract. Most gut bacterial modifications characterized so far are hydrolytic or reductive in nature. Here we report that abundant human gut bacteria from the phylum Bacteroidetes perform conjugative modifications by selectively sulfonating steroidal metabolites. While sulfonation is a ubiquitous biochemical modification, this activity has not yet been characterized in gut microbes. Using genetic and biochemical approaches, we identify a widespread biosynthetic gene cluster that encodes both a sulfotransferase (BtSULT, BT0416) and enzymes that synthesize the sulfonate donor adenosine 3'-phosphate-5'-phosphosulfate (PAPS), including an APS kinase (CysC, BT0413) and an ATP sulfurylase (CysD and CysN, BT0414-BT0415). BtSULT selectively sulfonates steroidal metabolites with a flat A/B ring fusion, including cholesterol. Germ-free mice monocolonized with Bacteroides thetaiotaomicron ΔBT0416 exhibited reduced gastrointestinal levels of cholesterol sulfate (Ch-S) compared with wild-type B. thetaiotaomicron-colonized mice. The presence of BtSULT and BtSULT homologues in bacteria inhibited leucocyte migration in vitro and in vivo, and abundances of cluster genes were significantly reduced in patients with inflammatory bowel disease. Together, these data provide a mechanism by which gut bacteria sulfonate steroidal metabolites and suggest that these compounds can modulate immune cell trafficking in the host.
Collapse
Affiliation(s)
- Lina Yao
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Gabriel D D'Agostino
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Jinseok Park
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Saiyu Hang
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Arijit A Adhikari
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Yancong Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Wei Li
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | | | - Sena Bae
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA.,Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eric A Franzosa
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA.,Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Curtis Huttenhower
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA.,Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - A Sloan Devlin
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Cholesterol sulfate alleviates ulcerative colitis by promoting cholesterol biosynthesis in colonic epithelial cells. Nat Commun 2022; 13:4428. [PMID: 35908039 PMCID: PMC9338998 DOI: 10.1038/s41467-022-32158-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 07/18/2022] [Indexed: 12/04/2022] Open
Abstract
Cholesterol sulfate, produced by hydroxysteroid sulfotransferase 2B1 (SULT2B1), is highly abundant in the intestine. Herein, we study the functional role and underlying intestinal epithelial repair mechanisms of cholesterol sulfate in ulcerative colitis. The levels of cholesterol and cholesterol sulfate, as well as the expression of Sult2b1 and genes involved in cholesterol biosynthesis, are significantly higher in inflamed tissues from patients with ulcerative colitis than in intestinal mucosa from healthy controls. Cholesterol sulfate in the gut and circulation is mainly catalyzed by intestinal epithelial SULT2B1. Specific deletion of the Sult2b1 gene in the intestinal epithelial cells aggravates dextran sulfate sodium-induced colitis; however, dietary supplementation with cholesterol sulfate ameliorates this effect in acute and chronic ulcerative colitis in mice. Cholesterol sulfate promotes cholesterol biosynthesis by binding to Niemann-Pick type C2 protein and activating sterol regulatory element binding protein 2 in colonic epithelial cells, thereby alleviates ulcerative colitis. In conclusion, cholesterol sulfate contributes to the healing of the mucosal barrier and exhibits therapeutic efficacy against ulcerative colitis in mice. New treatment strategies are required for ulcerative colitis. Here the authors show in mouse models that cholesterol sulfate, an endogenous active cholesterol derivative, contributes to the healing of the mucosal barrier by promoting cholesterol biosynthesis in colonic epithelial cells and exhibits therapeutic efficacy against ulcerative colitis.
Collapse
|
12
|
Dias IH, Shokr H, Shephard F, Chakrabarti L. Oxysterols and Oxysterol Sulfates in Alzheimer’s Disease Brain and Cerebrospinal Fluid. J Alzheimers Dis 2022; 87:1527-1536. [PMID: 35491790 PMCID: PMC9277668 DOI: 10.3233/jad-220083] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background: Brain cholesterol levels are tightly regulated but increasing evidence indicates that cholesterol metabolism may drive Alzheimer’s disease (AD)-associated pathological changes. Recent advances in understanding of mitochondrial dysfunction in AD brain have presented a vital role played by mitochondria in oxysterol biosynthesis and their involvement in pathophysiology. Oxysterol accumulation in brain is controlled by various enzymatic pathways including sulfation. While research into oxysterol is under the areas of active investigation, there is less evidence for oxysterol sulfate levels in human brain. Objective: This study investigates the hypothesis that AD brain oxysterol detoxification via sulfation is impaired in later stages of disease resulting in oxysterol accumulation. Methods: Lipids were extracted from postmortem frozen brain tissue and cerebrospinal (CSF) from late- (Braak stage III-IV) and early- (Braak stage I-II) stage AD patients. Samples were spiked with internal standards prior to lipid extraction. Oxysterols were enriched with a two-step solid phase extraction using a polymeric SPE column and further separation was achieved by LC-MS/MS. Results: Oxysterols, 26-hydroxycholesterol (26-OHC), 25-hydroxycholesterol (25-OHC), and 7-oxycholesterol levels were higher in brain tissue and mitochondria extracted from late-stage AD brain tissue except for 24S-hydroxycholesterol, which was decreased in late AD. However, oxysterol sulfates are significantly lower in the AD frontal cortex. Oxysterols, 25-OHC, and 7-oxocholesterol was higher is CSF but 26-OHC and oxysterol sulfate levels were not changed. Conclusion: Our results show oxysterol metabolism is altered in AD brain mitochondria, favoring synthesis of 26-OHC, 25-OHC, and 7-oxocholesterol, and this may influence brain mitochondrial function and acceleration of the disease.
Collapse
Affiliation(s)
- Irundika H.K. Dias
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Hala Shokr
- Manchester Pharmacy School, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Freya Shephard
- Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Lisa Chakrabarti
- Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
13
|
Thorne JL, Cioccoloni G. Nuclear Receptors and Lipid Sensing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:83-105. [DOI: 10.1007/978-3-031-11836-4_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
14
|
Sato K, Kakiyama G, Suzuki M, Naritaka N, Takei H, Sato H, Kimura A, Murai T, Kurosawa T, Pandak WM, Nittono H, Shimizu T. Changes in conjugated urinary bile acids across age groups. Steroids 2020; 164:108730. [PMID: 32961239 DOI: 10.1016/j.steroids.2020.108730] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/07/2020] [Accepted: 09/06/2020] [Indexed: 11/26/2022]
Abstract
Bile acid compositions are known to change dramatically after birth with aging. However, no reports have described the transition of conjugated urinary bile acids from the neonatal period to adulthood, and such findings would noninvasively offer insights into hepatic function. The aim of this study was to investigate differences in bile acid species, conjugation rates, and patterns, and to pool characteristics for age groups. We measured urinary bile acids in spot urine samples from 92 healthy individuals ranging from birth to 58 years old using liquid chromatography tandem mass spectrometry (LC/ESI-MS/MS). Sixty-six unconjugated and conjugated bile acids were systematically determined. After birth, urinary bile acids dramatically changed from fetal (i.e., Δ4-, Δ5-, and polyhydroxy-bile acids) to mature (i.e., CA and CDCA) bile acids. Peak bile acid excretion was 6-8 days after birth, steadily decreasing thereafter. A major change in bile acid conjugation pattern (taurine to glycine) also occurred at 2-4 months old. Our data provide important information regarding transitions of bile acid biosynthesis, including conjugation. The data also support the existence of physiologic cholestasis in the neonatal period and the establishment of the intestinal bacterial flora in infants.
Collapse
Affiliation(s)
- Keiko Sato
- Department of Pediatrics, Juntendo University, Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Genta Kakiyama
- Division of Gastroenterology, Hepatology, and Nutrition, Virginia Commonwealth University and McGuire VA Medical Center, 1201 Broad Rock Blvd., Richmond, VA 23249, USA.
| | - Mitsuyoshi Suzuki
- Department of Pediatrics, Juntendo University, Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Nakayuki Naritaka
- Junshin Clinic BA Institute, 2-1-22 Haramachi, Meguro-ku, Tokyo 152-0011, Japan.
| | - Hajime Takei
- Junshin Clinic BA Institute, 2-1-22 Haramachi, Meguro-ku, Tokyo 152-0011, Japan.
| | - Hiroaki Sato
- Department of Perinatal and Neonatal Medicine, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma-cho, Omiya-ku, Saitama 330-8503, Japan.
| | - Akihiko Kimura
- Department of Pediatrics and Child Health, Kurume University School of Medicine, 67 Asahi-cho, Kurume, Fukuoka 830-0011, Japan.
| | - Tsuyoshi Murai
- Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Tohbetsu-cho, Ishikari, Hokkaido 061-0293, Japan.
| | - Takao Kurosawa
- Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Tohbetsu-cho, Ishikari, Hokkaido 061-0293, Japan.
| | - William M Pandak
- Division of Gastroenterology, Hepatology, and Nutrition, Virginia Commonwealth University and McGuire VA Medical Center, 1201 Broad Rock Blvd., Richmond, VA 23249, USA.
| | - Hiroshi Nittono
- Junshin Clinic BA Institute, 2-1-22 Haramachi, Meguro-ku, Tokyo 152-0011, Japan.
| | - Toshiaki Shimizu
- Department of Pediatrics, Juntendo University, Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| |
Collapse
|
15
|
Sanchez LD, Pontini L, Marinozzi M, Sanchez-Aranguren LC, Reis A, Dias IHK. Cholesterol and oxysterol sulfates: Pathophysiological roles and analytical challenges. Br J Pharmacol 2020; 178:3327-3341. [PMID: 32762060 DOI: 10.1111/bph.15227] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/01/2020] [Accepted: 07/20/2020] [Indexed: 01/18/2023] Open
Abstract
Cholesterol and oxysterol sulfates are important regulators of lipid metabolism, inflammation, cell apoptosis, and cell survival. Among the sulfate-based lipids, cholesterol sulfate (CS) is the most studied lipid both quantitatively and functionally. Despite the importance, very few studies have analysed and linked the actions of oxysterol sulfates to their physiological and pathophysiological roles. Overexpression of sulfotransferases confirmed the formation of a range of oxysterol sulfates and their antagonistic effects on liver X receptors (LXRs) prompting further investigations how are the changes to oxysterol/oxysterol sulfate homeostasis can contribute to LXR activity in the physiological milieu. Here, we aim to bring together for novel roles of oxysterol sulfates, the available techniques and the challenges associated with their analysis. Understanding the oxysterol/oxysterol sulfate levels and their pathophysiological mechanisms could lead to new therapeutic targets for metabolic diseases. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
| | - Lorenzo Pontini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Maura Marinozzi
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | | | - Ana Reis
- LAQV/REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | | |
Collapse
|
16
|
Asghari A, Umetani M. Obesity and Cancer: 27-Hydroxycholesterol, the Missing Link. Int J Mol Sci 2020; 21:E4822. [PMID: 32650428 PMCID: PMC7404106 DOI: 10.3390/ijms21144822] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
Obesity is currently affecting more than 40% of the Americans, and if it progresses with this rate, soon one out of two Americans will be obese. Obesity is an important risk factor for several disorders including cardiovascular disease, the first cause of death in the United States. Cancer follows as the second deadliest disease, and a link between obesity and cancer has been suggested. However, it is very hard to establish an exact connection between obesity and cancers due to the multifactorial nature of obesity. Hypercholesterolemia is a comorbidity of obesity and also linked to several cancers. Recently a cholesterol metabolite 27-hydroxycholesterol (27HC) was found to be an endogenous selective estrogen receptor modulator (SERM), which opened new doors toward several interesting studies on the role of this molecule in biological disorders. It is speculated that 27HC might be the missing link in the obesity and cancer chain. Here, we explored the effects of 27-hydroxycholesterol on obesity and cancers with a focus on the SERM capacity of 27HC.
Collapse
Affiliation(s)
- Arvand Asghari
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204-5056, USA;
| | - Michihisa Umetani
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204-5056, USA;
- HEALTH Research Institute, University of Houston, Houston, TX 77204-5056, USA
| |
Collapse
|
17
|
Yi L, Dalai M, Su R, Lin W, Erdenedalai M, Luvsantseren B, Chimedtseren C, Wang Z, Hasi S. Whole-genome sequencing of wild Siberian musk deer (Moschus moschiferus) provides insights into its genetic features. BMC Genomics 2020; 21:108. [PMID: 32005147 PMCID: PMC6995116 DOI: 10.1186/s12864-020-6495-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/14/2020] [Indexed: 12/20/2022] Open
Abstract
Background Siberian musk deer, one of the seven species, is distributed in coniferous forests of Asia. Worldwide, the population size of Siberian musk deer is threatened by severe illegal poaching for commercially valuable musk and meat, habitat losses, and forest fire. At present, this species is categorized as Vulnerable on the IUCN Red List. However, the genetic information of Siberian musk deer is largely unexplored. Results Here, we produced 3.10 Gb draft assembly of wild Siberian musk deer with a contig N50 of 29,145 bp and a scaffold N50 of 7,955,248 bp. We annotated 19,363 protein-coding genes and estimated 44.44% of the genome to be repetitive. Our phylogenetic analysis reveals that wild Siberian musk deer is closer to Bovidae than to Cervidae. Comparative analyses showed that the genetic features of Siberian musk deer adapted in cold and high-altitude environments. We sequenced two additional genomes of Siberian musk deer constructed demographic history indicated that changes in effective population size corresponded with recent glacial epochs. Finally, we identified several candidate genes that may play a role in the musk secretion based on transcriptome analysis. Conclusions Here, we present a high-quality draft genome of wild Siberian musk deer, which will provide a valuable genetic resource for further investigations of this economically important musk deer.
Collapse
Affiliation(s)
- Li Yi
- Inner Mongolia Agricultural University / Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture and Rural Affairs, Hohhot, 010018, China
| | - Menggen Dalai
- Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China.
| | - Rina Su
- Inner Mongolia Agricultural University / Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture and Rural Affairs, Hohhot, 010018, China
| | - Weili Lin
- Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | | | | | | | - Zhen Wang
- Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Surong Hasi
- Inner Mongolia Agricultural University / Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture and Rural Affairs, Hohhot, 010018, China.
| |
Collapse
|
18
|
Anderson A, Campo A, Fulton E, Corwin A, Jerome WG, O'Connor MS. 7-Ketocholesterol in disease and aging. Redox Biol 2020; 29:101380. [PMID: 31926618 PMCID: PMC6926354 DOI: 10.1016/j.redox.2019.101380] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/06/2019] [Accepted: 11/10/2019] [Indexed: 02/08/2023] Open
Abstract
7-Ketocholesterol (7KC) is a toxic oxysterol that is associated with many diseases and disabilities of aging, as well as several orphan diseases. 7KC is the most common product of a reaction between cholesterol and oxygen radicals and is the most concentrated oxysterol found in the blood and arterial plaques of coronary artery disease patients as well as various other disease tissues and cell types. Unlike cholesterol, 7KC consistently shows cytotoxicity to cells and its physiological function in humans or other complex organisms is unknown. Oxysterols, particularly 7KC, have also been shown to diffuse through membranes where they affect receptor and enzymatic function. Here, we will explore the known and proposed mechanisms of pathologies that are associated with 7KC, as well speculate about the future of 7KC as a diagnostic and therapeutic target in medicine.
Collapse
|
19
|
Dias IH, Borah K, Amin B, Griffiths HR, Sassi K, Lizard G, Iriondo A, Martinez-Lage P. Localisation of oxysterols at the sub-cellular level and in biological fluids. J Steroid Biochem Mol Biol 2019; 193:105426. [PMID: 31301352 DOI: 10.1016/j.jsbmb.2019.105426] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/25/2019] [Accepted: 07/09/2019] [Indexed: 12/16/2022]
Abstract
Oxysterols are oxidized derivatives of cholesterol that are formed enzymatically or via reactive oxygen species or both. Cholesterol or oxysterols ingested as food are absorbed and packed into lipoproteins that are taken up by hepatic cells. Within hepatic cells, excess cholesterol is metabolised to form bile acids. The endoplasmic reticulum acts as the main organelle in the bile acid synthesis pathway. Metabolised sterols originating from this pathway are distributed within other organelles and in the cell membrane. The alterations to membrane oxysterol:sterol ratio affects the integrity of the cell membrane. The presence of oxysterols changes membrane fluidity and receptor orientation. It is well documented that hydroxylase enzymes located in mitochondria facilitate oxysterol production via an acidic pathway. More recently, the presence of oxysterols was also reported in lysosomes. Peroxisomal deficiencies favour intracellular oxysterols accumulation. Despite the low abundance of oxysterols compared to cholesterol, the biological actions of oxysterols are numerous and important. Oxysterol levels are implicated in the pathogenesis of multiple diseases ranging from chronic inflammatory diseases (atherosclerosis, Alzheimer's disease and bowel disease), cancer and numerous neurodegenerative diseases. In this article, we review the distribution of oxysterols in sub-cellular organelles and in biological fluids.
Collapse
Affiliation(s)
- Irundika Hk Dias
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, B4 7ET, UK.
| | - Khushboo Borah
- Faculty of Health and Medical Sciences, University of Surrey, Stag Hill, Guildford, GU2 7XH, UK
| | - Berivan Amin
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, B4 7ET, UK
| | - Helen R Griffiths
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, B4 7ET, UK; Faculty of Health and Medical Sciences, University of Surrey, Stag Hill, Guildford, GU2 7XH, UK
| | - Khouloud Sassi
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270)/University Bourgogne Franche-Comté/Inserm, 21000 Dijon, France; Univ. Tunis El Manar, Laboratory of Onco-Hematology (LR05ES05), Faculty of Medicine, Tunis, Tunisia
| | - Gérard Lizard
- Team Bio-PeroxIL, Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism (EA7270)/University Bourgogne Franche-Comté/Inserm, 21000 Dijon, France
| | - Ane Iriondo
- Department of Neurology, Center for Research and Advanced Therapies, CITA-Alzheimer Foundation, San Sebastian, Spain
| | - Pablo Martinez-Lage
- Department of Neurology, Center for Research and Advanced Therapies, CITA-Alzheimer Foundation, San Sebastian, Spain
| |
Collapse
|
20
|
Yang J, Broman MM, Cooper PO, Lanman NA, Strand DW, Morrissey C, Ratliff TL. Distinct expression patterns of SULT2B1b in human prostate epithelium. Prostate 2019; 79:1256-1266. [PMID: 31212370 PMCID: PMC7064034 DOI: 10.1002/pros.23829] [Citation(s) in RCA: 3] [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: 03/18/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND SULT2B1b (sulfotransferase family cytosolic 2B member 1b) catalyzes the sulfate conjugation of substrates such as cholesterol and oxysterols. Our laboratory has previously shown that SULT2B1b inhibition modulates androgen receptor signaling and induces apoptosis in prostate cancer cells. However, the functions of SULT2B1b in the prostate remain poorly understood. METHODS We characterized the expression pattern of SULT2B1b in human benign prostate hyperplasia (BPH) as well as prostate cancer to determine the relationship between SULT2B1b and prostate diseases, using immunohistochemistry, immunofluorescence staining, immunoblot, and real-time polymerase chain reaction. RESULTS SULT2B1b was strongly detected in the prostate epithelium but was absent in the stroma. Significantly lower SULT2B1b was found in primary cancer cells compared with adjacent normal epithelial cells. SULT2B1b further decreased in metastatic cancer cells. Most interestingly, we found, for the first time, that SULT2B1b was much more concentrated in the luminal layer than in the basal layer in both normal prostate and BPH samples. The stronger presence of SULT2B1b in luminal epithelial cells was confirmed by costaining with luminal and basal markers and in sorted paired luminal and basal cells. SULT2B1b expression was induced with prostate organoid differentiation. CONCLUSIONS SULT2B1b inversely correlates with prostate cancer status, with the highest level in the normal epithelium and lowest in the advanced metastatic prostate cancer. Furthermore, SULT2B1b is mostly located within the luminal layer of the prostate epithelium, suggesting that it may be implicated in luminal differentiation.
Collapse
Affiliation(s)
- Jiang Yang
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana
| | - Meaghan M. Broman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana
| | - Paula O. Cooper
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana
| | - Nadia A. Lanman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana
- Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Douglas W. Strand
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington
| | - Timothy L. Ratliff
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana
- Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana
| |
Collapse
|
21
|
Holy P, Kloudova A, Soucek P. Importance of genetic background of oxysterol signaling in cancer. Biochimie 2018; 153:109-138. [DOI: 10.1016/j.biochi.2018.04.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/27/2018] [Indexed: 12/14/2022]
|
22
|
Zimmer B, Tenbusch L, Klymiuk MC, Dezhkam Y, Schuler G. SULFATION PATHWAYS: Expression of SULT2A1, SULT2B1 and HSD3B1 in the porcine testis and epididymis. J Mol Endocrinol 2018; 61:M41-M55. [PMID: 29588428 DOI: 10.1530/jme-17-0277] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/27/2018] [Indexed: 01/01/2023]
Abstract
In the porcine testis, in addition to estrogen sulfates, the formation of numerous sulfonated neutral hydroxysteroids has been observed. However, their functions and the underlying synthetic pathways are still widely unclear. To obtain further information on their formation in postpubertal boars, the expression of sulfotransferases considered relevant for neutral hydroxysteroids (SULT2A1, SULT2B1) was investigated in the testis and defined segments of the epididymis applying real-time RT-qPCR, Western blot and immunohistochemistry (IHC). Sulfotransferase activities were assessed in tissue homogenates or cytosolic preparations applying dehydroepiandrosterone and pregnenolone as substrates. A high SULT2A1 expression was confirmed in the testis and localized in Leydig cells by IHC. In the epididymis, SULT2A1 expression was virtually confined to the body. SULT2B1 expression was absent or low in the testis but increased significantly along the epididymis. Immunohistochemical observations indicate that both enzymes are secreted into the ductal lumen via an apocrine mechanism. The results from the characterization of expression patterns and activity measurements suggest that SULT2A1 is the prevailing enzyme for the sulfonation of hydroxysteroids in the testis, whereas SULT2B1 may catalyze the formation of sterol sulfates in the epididymis. In order to obtain information on the overall steroidogenic capacity of the porcine epididymis, the expression of important steroidogenic enzymes (CYP11A1, CYP17A1, CYP19, HSD3B1, HSD17B3, SRD5A2) was monitored in the defined epididymal segments applying real-time RT-qPCR. Surprisingly, in addition to a high expression of SRD5A2 in the epididymal head, a substantial expression of HSD3B1 was detected, which increased along the organ.
Collapse
Affiliation(s)
- B Zimmer
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus-Liebig-University, Giessen, Germany
| | - L Tenbusch
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus-Liebig-University, Giessen, Germany
| | - M C Klymiuk
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus-Liebig-University, Giessen, Germany
| | - Y Dezhkam
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus-Liebig-University, Giessen, Germany
| | - G Schuler
- Veterinary Clinic for Obstetrics, Gynecology and Andrology, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
23
|
Laderoute H, Bone C, Squires EJ. The sulfoconjugation of androstenone and dehydroepiandrosterone by human and porcine sulfotransferase enzymes. Steroids 2018; 136:8-16. [PMID: 29792900 DOI: 10.1016/j.steroids.2018.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/26/2018] [Accepted: 05/14/2018] [Indexed: 10/16/2022]
Abstract
Porcine sulfotransferase 2A1 (pSULT2A1) is a key enzyme involved in the testicular and hepatic sulfoconjugation of steroids such as dehydroepiandrosterone (DHEA) and potentially androstenone. This latter steroid is a major cause of boar taint, which is an unpleasant off-odour and off-flavour in pork from male pigs. Sulfotransferase 2B1 (pSULT2B1) may also be important, although no direct evidence exists for its involvement in sulfoconjugation of steroids. The purpose of this study was to investigate the sulfoconjugation activity of human and porcine sulfotransferases towards DHEA and androstenone. pcDNA 3.1 vectors expressing porcine (p) SULT2A1, pSULT2B1, human (h) SULT2A1, hSULT2B1a, and hSULT2B1b enzymes were transfected into human embryonic kidney cells. Transfected cells were then incubated with either androstenone or dehydroepiandrosterone (DHEA) in both time-course and enzyme kinetics studies. The production of sulfonates of androstenone metabolites and DHEA sulfonate increased over time for all enzymes with the exception of pSULT2B1. Enzyme kinetics analysis showed that androstenone and DHEA were poor substrates for the human orthologs, hSULT2B1a and hSULT2B1b. Human and porcine SULT2A1 showed substantially different substrate affinities for androstenone (Km 5.8 ± 0.6 µM and 74.1 ± 15.9 µM, respectively) and DHEA (Km 9.4 ± 2.5 µM and 3.3 ± 1.9 µM, respectively). However, these enzymes did show relatively similar sulfonation efficiencies for DHEA (Vmax/Km 50.5 and 72.9 for hSULT2A1 and pSULT2A1, respectively). These results highlight the species differences in sulfonation activity and provide direct evidence, for the first time, suggesting that pSULT2B1 is not involved in sulfonation of either androstenone metabolites or DHEA.
Collapse
Affiliation(s)
- Heidi Laderoute
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario N1G2W1, Canada
| | - Christine Bone
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario N1G2W1, Canada
| | - E James Squires
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario N1G2W1, Canada.
| |
Collapse
|
24
|
Sakurai T, Uruno T, Sugiura Y, Tatsuguchi T, Yamamura K, Ushijima M, Hattori Y, Kukimoto-Niino M, Mishima-Tsumagari C, Watanabe M, Suematsu M, Fukui Y. Cholesterol sulfate is a DOCK2 inhibitor that mediates tissue-specific immune evasion in the eye. Sci Signal 2018; 11:11/541/eaao4874. [DOI: 10.1126/scisignal.aao4874] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
25
|
Gallo C, Nuzzo G, d'Ippolito G, Manzo E, Sardo A, Fontana A. Sterol Sulfates and Sulfotransferases in Marine Diatoms. Methods Enzymol 2018; 605:101-138. [PMID: 29909823 DOI: 10.1016/bs.mie.2018.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sterol sulfates are widely occurring molecules in marine organisms. Their importance has been so far underestimated although many of these compounds are crucial mediators of physiological and ecological functions in other organisms. Biosynthesis of sterol sulfates is controlled by cytosolic sulfotransferases (SULTs), a varied family of enzymes that catalyze the transfer of a sulfo residue (-SO3H) from the universal donor 3'-phosphoadenosine-5'-phosphosulfate to the hydroxyl function at C-3 of the steroid skeleton. The absence of molecular tools has been the main impediment to the development of a biosynthetic study of this class of compounds in marine organisms. In fact, there is very limited information about these enzymes in marine environments. SULT activity has, however, been reported in several marine species, and, recently, the production of sterol sulfates has been linked to the control of growth in marine diatoms. In this chapter, we describe methods for the study of sterol sulfates in this lineage of marine microalgae. The main aim is to provide the tools useful to deal with the biosynthesis and regulation of these compounds and to circumvent the bottleneck of the lack of molecular information. The protocols have been designed for marine diatoms, but most of the procedures can be used for other marine organisms.
Collapse
Affiliation(s)
- Carmela Gallo
- National Research Council of Italy, Institute of Biomolecular Chemistry, Pozzuoli, Naples, Italy
| | - Genoveffa Nuzzo
- National Research Council of Italy, Institute of Biomolecular Chemistry, Pozzuoli, Naples, Italy
| | - Giuliana d'Ippolito
- National Research Council of Italy, Institute of Biomolecular Chemistry, Pozzuoli, Naples, Italy.
| | - Emiliano Manzo
- National Research Council of Italy, Institute of Biomolecular Chemistry, Pozzuoli, Naples, Italy
| | - Angela Sardo
- National Research Council of Italy, Institute of Biomolecular Chemistry, Pozzuoli, Naples, Italy
| | - Angelo Fontana
- National Research Council of Italy, Institute of Biomolecular Chemistry, Pozzuoli, Naples, Italy.
| |
Collapse
|
26
|
Autoinhibitory sterol sulfates mediate programmed cell death in a bloom-forming marine diatom. Nat Commun 2017; 8:1292. [PMID: 29101388 PMCID: PMC5670183 DOI: 10.1038/s41467-017-01300-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 09/07/2017] [Indexed: 01/07/2023] Open
Abstract
Cell mortality is a key mechanism that shapes phytoplankton blooms and species dynamics in aquatic environments. Here we show that sterol sulfates (StS) are regulatory molecules of a cell death program in Skeletonema marinoi, a marine diatom-blooming species in temperate coastal waters. The molecules trigger an oxidative burst and production of nitric oxide in a dose-dependent manner. The intracellular level of StS increases with cell ageing and ultimately leads to a mechanism of apoptosis-like death. Disrupting StS biosynthesis by inhibition of the sulfonation step significantly delays the onset of this fatal process and maintains steady growth in algal cells for several days. The autoinhibitory activity of StS demonstrates the functional significance of small metabolites in diatoms. The StS pathway provides another view on cell regulation during bloom dynamics in marine habitats and opens new opportunities for the biochemical control of mass-cultivation of microalgae. Phytoplankton blooms are shaped by a period of rapid growth followed by massive cell death. Here the authors show that sterol sulfates accumulate in aging cells of a bloom-forming marine diatom and trigger an oxidative burst that leads to a mechanism of apoptosis-like death.
Collapse
|
27
|
Johnson CH, Santidrian AF, LeBoeuf SE, Kurczy ME, Rattray NJW, Rattray Z, Warth B, Ritland M, Hoang LT, Loriot C, Higa J, Hansen JE, Felding BH, Siuzdak G. Metabolomics guided pathway analysis reveals link between cancer metastasis, cholesterol sulfate, and phospholipids. Cancer Metab 2017; 5:9. [PMID: 29093815 PMCID: PMC5663111 DOI: 10.1186/s40170-017-0171-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 10/06/2017] [Indexed: 01/02/2023] Open
Abstract
Background Cancer cells that enter the metastatic cascade require traits that allow them to survive within the circulation and colonize distant organ sites. As disseminating cancer cells adapt to their changing microenvironments, they also modify their metabolism and metabolite production. Methods A mouse xenograft model of spontaneous tumor metastasis was used to determine the metabolic rewiring that occurs between primary cancers and their metastases. An “autonomous” mass spectrometry-based untargeted metabolomic workflow with integrative metabolic pathway analysis revealed a number of differentially regulated metabolites in primary mammary fat pad (MFP) tumors compared to microdissected paired lung metastases. The study was further extended to analyze metabolites in paired normal tissues which determined the potential influence of metabolites from the microenvironment. Results Metabolomic analysis revealed that multiple metabolites were increased in metastases, including cholesterol sulfate and phospholipids (phosphatidylglycerols and phosphatidylethanolamine). Metabolite analysis of normal lung tissue in the mouse model also revealed increased levels of these metabolites compared to tissues from normal MFP and primary MFP tumors, indicating potential extracellular uptake by cancer cells in lung metastases. These results indicate a potential functional importance of cholesterol sulfate and phospholipids in propagating metastasis. In addition, metabolites involved in DNA/RNA synthesis and the TCA cycle were decreased in lung metastases compared to primary MFP tumors. Conclusions Using an integrated metabolomic workflow, this study identified a link between cholesterol sulfate and phospholipids, metabolic characteristics of the metastatic niche, and the capacity of tumor cells to colonize distant sites. Electronic supplementary material The online version of this article (10.1186/s40170-017-0171-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Caroline H Johnson
- Scripps Center for Metabolomics, The Scripps Research Institute, La Jolla, CA USA.,Department of Environmental Health Sciences, Yale School of Public HealthYale School of Medicine, New Haven, CT USA.,Yale Cancer Center, Yale School of Medicine, New Haven, CT USA
| | - Antonio F Santidrian
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA USA.,Current address: Department of Molecular Oncology and Immunotherapies, StemImmune, Inc., San Diego, CA 92122 USA
| | - Sarah E LeBoeuf
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA USA.,Current address: NYU Langone Medical Center, New York, NY 10016 USA
| | - Michael E Kurczy
- Scripps Center for Metabolomics, The Scripps Research Institute, La Jolla, CA USA.,Current address: CVMD IMED AstraZeneca, Gothenburg, Sweden
| | - Nicholas J W Rattray
- Department of Environmental Health Sciences, Yale School of Public HealthYale School of Medicine, New Haven, CT USA
| | - Zahra Rattray
- Department of Therapeutic Radiology, Yale School of Medicine, Yale University, New Haven, CT 06520 USA
| | - Benedikt Warth
- Scripps Center for Metabolomics, The Scripps Research Institute, La Jolla, CA USA
| | - Melissa Ritland
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA USA.,Current address: Active Motif Inc, Carlsbad, CA 92008 USA
| | - Linh T Hoang
- Scripps Center for Metabolomics, The Scripps Research Institute, La Jolla, CA USA
| | - Celine Loriot
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA USA
| | - Jason Higa
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA USA
| | - James E Hansen
- Yale Cancer Center, Yale School of Medicine, New Haven, CT USA.,Department of Therapeutic Radiology, Yale School of Medicine, Yale University, New Haven, CT 06520 USA
| | - Brunhilde H Felding
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA USA
| | - Gary Siuzdak
- Scripps Center for Metabolomics, The Scripps Research Institute, La Jolla, CA USA
| |
Collapse
|
28
|
Heinz L, Kim GJ, Marrakchi S, Christiansen J, Turki H, Rauschendorf MA, Lathrop M, Hausser I, Zimmer AD, Fischer J. Mutations in SULT2B1 Cause Autosomal-Recessive Congenital Ichthyosis in Humans. Am J Hum Genet 2017; 100:926-939. [PMID: 28575648 DOI: 10.1016/j.ajhg.2017.05.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 05/05/2017] [Indexed: 12/30/2022] Open
Abstract
Ichthyoses are a clinically and genetically heterogeneous group of genodermatoses associated with abnormal scaling of the skin over the whole body. Mutations in nine genes are known to cause non-syndromic forms of autosomal-recessive congenital ichthyosis (ARCI). However, not all genetic causes for ARCI have been discovered to date. Using whole-exome sequencing (WES) and multigene panel screening, we identified 6 ARCI-affected individuals from three unrelated families with mutations in Sulfotransferase family 2B member 1 (SULT2B1), showing their causative association with ARCI. Cytosolic sulfotransferases form a large family of enzymes that are involved in the synthesis and metabolism of several steroids in humans. We identified four distinct mutations including missense, nonsense, and splice site mutations. We demonstrated the loss of SULT2B1 expression at RNA and protein levels in keratinocytes from individuals with ARCI by functional analyses. Furthermore, we succeeded in reconstructing the morphologic skin alterations in a 3D organotypic tissue culture model with SULT2B1-deficient keratinocytes and fibroblasts. By thin layer chromatography (TLC) of extracts from these organotypic cultures, we could show the absence of cholesterol sulfate, the metabolite of SULT2B1, and an increased level of cholesterol, indicating a disturbed cholesterol metabolism of the skin upon loss-of-function mutation in SULT2B1. In conclusion, our study reveals an essential role for SULT2B1 in the proper development of healthy human skin. Mutation in SULT2B1 leads to an ARCI phenotype via increased proliferation of human keratinocytes, thickening of epithelial layers, and altered epidermal cholesterol metabolism.
Collapse
|
29
|
Ning Y, Kim JK, Min HK, Ren S. Cholesterol metabolites alleviate injured liver function and decrease mortality in an LPS-induced mouse model. Metabolism 2017; 71:83-93. [PMID: 28521882 DOI: 10.1016/j.metabol.2016.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 12/01/2016] [Accepted: 12/07/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Oxysterol sulfation plays a fundamental role in the regulation of many biological events. Its products, 25-hydroxycholesterol 3-sulfate (25HC3S) and 25-hydroxycholesterol 3, 25-disulfate (25HCDS), have been demonstrated to be potent regulators of lipid metabolism, inflammatory response, cell apoptosis, and cell survival. In the present study, we tested these products' potential to treat LPS-induced acute liver failure in a mouse model. METHODS Acute liver failure mouse model was established by intravenous injection with LPS. The injured liver function was treated with intraperitoneal administration of 25HC, 25HC3S or 25HCDS. Serum enzymatic activities were determined in our clinic laboratory. ELISA assays were used to detect pro-inflammatory factor levels in sera. Western blot, Real-time Quantitative PCR and RT2 Profiler PCR Array analysis were used to determine levels of gene expression. RESULTS Administration of 25HC3S/25HCDS decreased serum liver-impaired markers; suppressed secretion of pro-inflammatory factors; alleviated liver, lung, and kidney injury; and subsequently increased the survival rate in the LPS-induced mouse model. These effects resulted from the inhibition of the expression of genes involved in the pro-inflammatory response and apoptosis and the simultaneous induction of the expression of genes involved in cell survival. Compared to 25HC, 25HC3S and 25HCDS exhibited significantly stronger effects in these activities, indicating that the cholesterol metabolites play an important role in the inflammatory response, cell apoptosis, and cell survival in vivo. CONCLUSIONS 25HC3S/25HCDS has potential to serve as novel biomedicines in the therapy of acute liver failure and acute multiple organ failure.
Collapse
Affiliation(s)
- Yanxia Ning
- Department of Internal Medicine, Virginia Commonwealth University/McGuire Veterans Affairs Medical Center, Richmond, VA 23249, United States
| | - Jin Kyung Kim
- Department of Internal Medicine, Virginia Commonwealth University/McGuire Veterans Affairs Medical Center, Richmond, VA 23249, United States
| | - Hae-Ki Min
- Department of Internal Medicine, Virginia Commonwealth University/McGuire Veterans Affairs Medical Center, Richmond, VA 23249, United States
| | - Shunlin Ren
- Department of Internal Medicine, Virginia Commonwealth University/McGuire Veterans Affairs Medical Center, Richmond, VA 23249, United States.
| |
Collapse
|
30
|
Rasool MI, Bairam AF, Kurogi K, Liu MC. On the sulfation of O-desmethyltramadol by human cytosolic sulfotransferases. Pharmacol Rep 2017; 69:953-958. [PMID: 28802998 DOI: 10.1016/j.pharep.2017.02.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/01/2017] [Accepted: 02/14/2017] [Indexed: 01/20/2023]
Abstract
BACKGROUND Previous studies have demonstrated that sulfate conjugation is involved in the metabolism of the active metabolite of tramadol, O-desmethyltramadol (O-DMT). The current study aimed to systematically identify the human cytosolic sulfotransferases (SULTs) that are capable of mediating the sulfation of O-DMT. METHODS The sulfation of O-DMT under metabolic conditions was demonstrated using HepG2 hepatoma cells and Caco-2 human colon carcinoma cells. O-DMT-sulfating activity of thirteen known human SULTs and four human organ specimens was examined using an established sulfotransferase assay. pH-Dependency and kinetic parameters were also analyzed using, respectively, buffers at different pHs and varying O-DMT concentrations in the assays. RESULTS Of the thirteen human SULTs tested, only SULT1A3 and SULT1C4 were found to display O-DMT-sulfating activity, with different pH-dependency profiles. Kinetic analysis revealed that SULT1C4 was 60 times more catalytically efficient in mediating the sulfation of O-DMT than SULT1A3 at respective optimal pH. Of the four human organ specimens tested, the cytosol prepared from the small intestine showed much higher O-DMT-sulfating activity than cytosols prepared from liver, lung, and kidney. Both cultured HepG2 and Caco-2 cells were shown to be capable of sulfating O-DMT and releasing sulfated O-DMT into cultured media. CONCLUSION SULT1A3 and SULT1C4 were the major SULTs responsible for the sulfation of O-DMT. Collectively, the results obtained provided a molecular basis underlying the sulfation of O-DMT and contributed to a better understanding about the pharmacokinetics and pharmacodynamics of tramadol in humans.
Collapse
Affiliation(s)
- Mohammed I Rasool
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, USA; Department of Pharmacology and Toxicology, College of Pharmacy, University of Karbala, Karbala, Iraq
| | - Ahsan F Bairam
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, USA; Department of Pharmacology and Toxicology, College of Pharmacy, University of Kufa, Kufa, Iraq
| | - Katsuhisa Kurogi
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, USA; Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki, Japan
| | - Ming-Cheh Liu
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, USA.
| |
Collapse
|
31
|
Bairam AF, Rasool MI, Kurogi K, Liu MC. On the Molecular Basis Underlying the Metabolism of Tapentadol Through Sulfation. Eur J Drug Metab Pharmacokinet 2017; 42:793-800. [DOI: 10.1007/s13318-016-0392-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
32
|
Mutemberezi V, Guillemot-Legris O, Muccioli GG. Oxysterols: From cholesterol metabolites to key mediators. Prog Lipid Res 2016; 64:152-169. [PMID: 27687912 DOI: 10.1016/j.plipres.2016.09.002] [Citation(s) in RCA: 246] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/13/2016] [Accepted: 09/23/2016] [Indexed: 12/13/2022]
Abstract
Oxysterols are cholesterol metabolites that can be produced through enzymatic or radical processes. They constitute a large family of lipids (i.e. the oxysterome) involved in a plethora of physiological processes. They can act through GPCR (e.g. EBI2, SMO, CXCR2), nuclear receptors (LXR, ROR, ERα) and through transporters or regulatory proteins. Their physiological effects encompass cholesterol, lipid and glucose homeostasis. Additionally, they were shown to be involved in other processes such as immune regulatory functions and brain homeostasis. First studied as precursors of bile acids, they quickly emerged as interesting lipid mediators. Their levels are greatly altered in several pathologies and some oxysterols (e.g. 4β-hydroxycholesterol or 7α-hydroxycholestenone) are used as biomarkers of specific pathologies. In this review, we discuss the complex metabolism and molecular targets (including binding properties) of these bioactive lipids in human and mice. We also discuss the genetic mouse models currently available to interrogate their effects in pathophysiological settings. We also summarize the levels of oxysterols reported in two key organs in oxysterol metabolism (liver and brain), plasma and cerebrospinal fluid. Finally, we consider future opportunities and directions in the oxysterol field in order to gain a better insight and understanding of the complex oxysterol system.
Collapse
Affiliation(s)
- Valentin Mutemberezi
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium
| | - Owein Guillemot-Legris
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Belgium.
| |
Collapse
|
33
|
Integrated application of transcriptomics and metabolomics yields insights into population-asynchronous ovary development in Coilia nasus. Sci Rep 2016; 6:31835. [PMID: 27545088 PMCID: PMC4992829 DOI: 10.1038/srep31835] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/27/2016] [Indexed: 11/11/2022] Open
Abstract
Populations of Coilia nasus demonstrate asynchronous ovarian development, which severely restricts artificial breeding and large-scale cultivation. In this study, we used a combination of transcriptomic and metabolomic methods to identify the key signaling pathways and genes regulation affecting ovarian development. We identified 565 compounds and generated 47,049 unigenes from ovary tissue. Fifteen metabolites and 830 genes were significantly up-regulated, while 27 metabolites and 642 genes were significantly down-regulated from stage III to stage IV of ovary development. Meanwhile, 31 metabolites and 1,932 genes were significantly up-regulated, and four metabolites and 764 genes were down-regulated from stage IV to stage V. These differentially expressed genes and metabolites were enriched by MetScape. Forty-three and 50 signaling pathways had important functions from stage III–IV and from stage IV–V in the ovary, respectively. Among the above signaling pathways, 39 played important roles from ovarian stage III–V, including “squalene and cholesterol biosynthesis”, “steroid hormone biosynthesis”, and “arachidonate metabolism and prostaglandin formation” pathways which may thus have key roles in regulating asynchronous development. These results shed new light on our understanding of the mechanisms responsible for population-asynchronous development in fish.
Collapse
|
34
|
Coughtrie MWH. Function and organization of the human cytosolic sulfotransferase (SULT) family. Chem Biol Interact 2016; 259:2-7. [PMID: 27174136 DOI: 10.1016/j.cbi.2016.05.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/02/2016] [Indexed: 12/29/2022]
Abstract
The sulfuryl transfer reaction is of fundamental biological importance. One of the most important manifestations of this process are the reactions catalyzed by members of the cytosolic sulfotransferase (SULT) superfamily. These enzymes transfer the sulfuryl moiety from the universal donor PAPS (3'-phosphoadenosine 5'-phosphosulfate) to a wide variety of substrates with hydroxyl- or amino-groups. Normally a detoxification reaction this facilitates the elimination of a multitude of xenobiotics, although for some molecules sulfation is a bioactivation step. In addition, sulfation plays a key role in endocrine and other signalling pathways since many steroids, sterols, thyroid hormones and catecholamines exist primarily as sulfate conjugates in humans. This article summarizes much of our current knowledge of the organization and function of the human cytosolic sulfotransferases and highlights some of the important interspecies differences that have implications for, among other things, drug development and chemical safety analysis.
Collapse
Affiliation(s)
- Michael W H Coughtrie
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
35
|
Guo Y, Hu B, Xie Y, Billiar TR, Sperry JL, Huang M, Xie W. Regulation of drug-metabolizing enzymes by local and systemic liver injuries. Expert Opin Drug Metab Toxicol 2016; 12:245-51. [PMID: 26751558 DOI: 10.1517/17425255.2016.1139574] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Drug metabolism and disposition are critical in maintaining the chemical and functional homeostasis of xenobiotics/drugs and endobiotics. The liver plays an essential role in drug metabolism and disposition due to its abundant expression of drug-metabolizing enzymes (DMEs) and transporters. There is growing evidence to suggest that many hepatic and systemic diseases can affect drug metabolism and disposition by regulating the expression and/or activity of DMEs and transporters in the liver. AREAS COVERED This review focuses on the recent progress on the regulation of DMEs by local and systemic liver injuries. Liver ischemia and reperfusion (I/R) and sepsis are used as examples of local and systemic injury, respectively. The reciprocal effect of the expression and activity of DMEs on animals' sensitivity to local and systemic liver injuries is also discussed. EXPERT OPINION Local and systemic liver injuries have a major effect on the expression and activity of DMEs in the liver. Understanding the disease effect on DMEs is clinically important due to the concern of disease-drug interactions. Future studies are necessary to understand the mechanism by which liver injury regulates DMEs. Human studies are also urgently needed in order to determine whether the results in animals can be replicated in human patients.
Collapse
Affiliation(s)
- Yan Guo
- a Center for Pharmacogenetics and Department of Pharmaceutical Sciences , University of Pittsburgh , Pittsburgh , PA , USA.,b Department of Pathology , Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Bingfang Hu
- a Center for Pharmacogenetics and Department of Pharmaceutical Sciences , University of Pittsburgh , Pittsburgh , PA , USA.,c Institute of Clinical Pharmacology , Sun Yat-Sen University , Guangzhou , China
| | - Yang Xie
- a Center for Pharmacogenetics and Department of Pharmaceutical Sciences , University of Pittsburgh , Pittsburgh , PA , USA
| | - Timothy R Billiar
- d Department of Surgery , University of Pittsburgh , Pittsburgh , PA , USA
| | - Jason L Sperry
- d Department of Surgery , University of Pittsburgh , Pittsburgh , PA , USA
| | - Min Huang
- c Institute of Clinical Pharmacology , Sun Yat-Sen University , Guangzhou , China
| | - Wen Xie
- a Center for Pharmacogenetics and Department of Pharmaceutical Sciences , University of Pittsburgh , Pittsburgh , PA , USA
| |
Collapse
|
36
|
VAŇKOVÁ M, HILL M, VELÍKOVÁ M, VČELÁK J, VACÍNOVÁ G, LUKÁŠOVÁ P, VEJRAŽKOVÁ D, DVOŘÁKOVÁ K, RUSINA R, HOLMEROVÁ I, JAROLÍMOVÁ E, VAŇKOVÁ H, BENDLOVÁ B. Reduced Sulfotransferase SULT2A1 Activity in Patients With Alzheimer´s Disease. Physiol Res 2015; 64:S265-73. [DOI: 10.33549/physiolres.933160] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Steroids are important components in the pathophysiology of Alzheimer’s disease (AD). Although their role has been studied, the corresponding metabolomic data is limited. In the present study we evaluate the role of steroid sulfotransferase SULT2A1 in the pathophysiology of AD on the basis of circulating steroids (measured by GC-MS), in which the sulfation catalyzed by SULT2A1 dominates over glucuronidation (pregnenolone/sulfate, DHEA/sulfate, androstenediol/sulfate and 5α-reduced pregnane and androstane catabolites). To estimate a general trend of SUL2A1 activity in AD patients we compared the ratios of steroid conjugates to their unconjugated counterparts (C/U) in controls (11 men and 22 women) and AD patients (18 men and 16 women) for individual circulating steroids after adjustment for age and BMI using ANCOVA model including the factors AD status and gender. Decreased C/U ratio for the C19 steroids demonstrate an association between attenuated sulfation of C19 steroids in adrenal zona reticularis and the pathophysiology of AD.
Collapse
Affiliation(s)
- M. VAŇKOVÁ
- Institute of Endocrinology, Prague, Czech Republic
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Mueller JW, Gilligan LC, Idkowiak J, Arlt W, Foster PA. The Regulation of Steroid Action by Sulfation and Desulfation. Endocr Rev 2015; 36:526-63. [PMID: 26213785 PMCID: PMC4591525 DOI: 10.1210/er.2015-1036] [Citation(s) in RCA: 311] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 07/21/2015] [Indexed: 12/14/2022]
Abstract
Steroid sulfation and desulfation are fundamental pathways vital for a functional vertebrate endocrine system. After biosynthesis, hydrophobic steroids are sulfated to expedite circulatory transit. Target cells express transmembrane organic anion-transporting polypeptides that facilitate cellular uptake of sulfated steroids. Once intracellular, sulfatases hydrolyze these steroid sulfate esters to their unconjugated, and usually active, forms. Because most steroids can be sulfated, including cholesterol, pregnenolone, dehydroepiandrosterone, and estrone, understanding the function, tissue distribution, and regulation of sulfation and desulfation processes provides significant insights into normal endocrine function. Not surprisingly, dysregulation of these pathways is associated with numerous pathologies, including steroid-dependent cancers, polycystic ovary syndrome, and X-linked ichthyosis. Here we provide a comprehensive examination of our current knowledge of endocrine-related sulfation and desulfation pathways. We describe the interplay between sulfatases and sulfotransferases, showing how their expression and regulation influences steroid action. Furthermore, we address the role that organic anion-transporting polypeptides play in regulating intracellular steroid concentrations and how their expression patterns influence many pathologies, especially cancer. Finally, the recent advances in pharmacologically targeting steroidogenic pathways will be examined.
Collapse
Affiliation(s)
- Jonathan W Mueller
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Lorna C Gilligan
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Jan Idkowiak
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Wiebke Arlt
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Paul A Foster
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
38
|
Xu Z, Gan L, Li T, Xu C, Chen K, Wang X, Qin JG, Chen L, Li E. Transcriptome Profiling and Molecular Pathway Analysis of Genes in Association with Salinity Adaptation in Nile Tilapia Oreochromis niloticus. PLoS One 2015; 10:e0136506. [PMID: 26305564 PMCID: PMC4548949 DOI: 10.1371/journal.pone.0136506] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/04/2015] [Indexed: 12/14/2022] Open
Abstract
Nile tilapia Oreochromis niloticus is a freshwater fish but can tolerate a wide range of salinities. The mechanism of salinity adaptation at the molecular level was studied using RNA-Seq to explore the molecular pathways in fish exposed to 0, 8, or 16 (practical salinity unit, psu). Based on the change of gene expressions, the differential genes unions from freshwater to saline water were classified into three categories. In the constant change category (1), steroid biosynthesis, steroid hormone biosynthesis, fat digestion and absorption, complement and coagulation cascades were significantly affected by salinity indicating the pivotal roles of sterol-related pathways in response to salinity stress. In the change-then-stable category (2), ribosomes, oxidative phosphorylation, signaling pathways for peroxisome proliferator activated receptors, and fat digestion and absorption changed significantly with increasing salinity, showing sensitivity to salinity variation in the environment and a responding threshold to salinity change. In the stable-then-change category (3), protein export, protein processing in endoplasmic reticulum, tight junction, thyroid hormone synthesis, antigen processing and presentation, glycolysis/gluconeogenesis and glycosaminoglycan biosynthesis—keratan sulfate were the significantly changed pathways, suggesting that these pathways were less sensitive to salinity variation. This study reveals fundamental mechanism of the molecular response to salinity adaptation in O. niloticus, and provides a general guidance to understand saline acclimation in O. niloticus.
Collapse
Affiliation(s)
- Zhixin Xu
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Rd., Shanghai 200241, China
| | - Lei Gan
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Rd., Shanghai 200241, China
| | - Tongyu Li
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Rd., Shanghai 200241, China
| | - Chang Xu
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Rd., Shanghai 200241, China
| | - Ke Chen
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Rd., Shanghai 200241, China
| | - Xiaodan Wang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Rd., Shanghai 200241, China
| | - Jian G. Qin
- School of Biological Sciences, Flinders University, Adelaide, SA 5001, Australia
| | - Liqiao Chen
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Rd., Shanghai 200241, China
- * E-mail: (EL); (LC)
| | - Erchao Li
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, 500 Dongchuan Rd., Shanghai 200241, China
- * E-mail: (EL); (LC)
| |
Collapse
|
39
|
Chen BH, Wang CC, Hou YH, Mao YC, Yang YS. Mechanism of sulfotransferase pharmacogenetics in altered xenobiotic metabolism. Expert Opin Drug Metab Toxicol 2015; 11:1053-71. [DOI: 10.1517/17425255.2015.1045486] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
40
|
Yamamoto A, Liu MY, Kurogi K, Sakakibara Y, Saeki Y, Suiko M, Liu MC. Sulphation of acetaminophen by the human cytosolic sulfotransferases: a systematic analysis. J Biochem 2015; 158:497-504. [PMID: 26067475 DOI: 10.1093/jb/mvv062] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 05/26/2015] [Indexed: 12/16/2022] Open
Abstract
Sulphation is known to be critically involved in the metabolism of acetaminophen in vivo. This study aimed to systematically identify the major human cytosolic sulfotransferase (SULT) enzyme(s) responsible for the sulphation of acetaminophen. A systematic analysis showed that three of the twelve human SULTs, SULT1A1, SULT1A3 and SULT1C4, displayed the strongest sulphating activity towards acetaminophen. The pH dependence of the sulphation of acetaminophen by each of these three SULTs was examined. Kinetic parameters of these three SULTs in catalysing acetaminophen sulphation were determined. Moreover, sulphation of acetaminophen was shown to occur in HepG2 human hepatoma cells and Caco-2 human intestinal epithelial cells under the metabolic setting. Of the four human organ samples tested, liver and intestine cytosols displayed considerably higher acetaminophen-sulphating activity than those of lung and kidney. Collectively, these results provided useful information concerning the biochemical basis underlying the metabolism of acetaminophen in vivo previously reported.
Collapse
Affiliation(s)
- Akihiro Yamamoto
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614, USA; Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan; and
| | - Ming-Yih Liu
- National Synchrotron Radiation Research Center, Hsinchu 30076, Taiwan
| | - Katsuhisa Kurogi
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614, USA; Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan; and
| | - Yoichi Sakakibara
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan; and
| | - Yuichi Saeki
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan; and
| | - Masahito Suiko
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan; and
| | - Ming-Cheh Liu
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614, USA;
| |
Collapse
|
41
|
Bellezza I, Gatticchi L, del Sordo R, Peirce MJ, Sidoni A, Roberti R, Minelli A. The loss of Tm7sf gene accelerates skin papilloma formation in mice. Sci Rep 2015; 5:9471. [PMID: 25804527 PMCID: PMC4372794 DOI: 10.1038/srep09471] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 02/16/2015] [Indexed: 01/04/2023] Open
Abstract
The 3β-hydroxysterol Δ14-reductase, encoded by the Tm7sf2 gene, is an enzyme involved in cholesterol biosynthesis. Cholesterol and its derivatives control epidermal barrier integrity and are protective against environmental insults. To determine the role of the gene in skin cholesterol homeostasis, we applied 12-o-tetradecanoylphorbol-13-acetate (TPA) to the skin of Tm7sf2+/+ and Tm7sf2-/- mice. TPA increased skin cholesterol levels by inducing de novo synthesis and up-take only in Tm7sf2+/+ mouse, confirming that the gene maintains cholesterol homeostasis under stress conditions. Cholesterol sulfate, one of the major players in skin permeability, was doubled by TPA treatment in the skin of wild-type animals but this response was lost in Tm7sf2-/- mice. The expression of markers of epidermal differentiation concomitant with farnesoid-X-receptor and p38 MAPK activation were also disrupted in Tm7sf2-/- mice. We then subjected Tm7sf2+/+ and Tm7sf2-/- mice to a classical two-stage skin carcinogenesis protocol. We found that the loss of Tm7sf2 increased incidence and multiplicity of skin papillomas. Interestingly, the null genotype showed reduced expression of nur77, a gene associated with resistance to neoplastic transformation. In conclusion, the loss of Tm7sf2 alters the expression of proteins involved in epidermal differentiation by reducing the levels of cholesterol sulfate.
Collapse
Affiliation(s)
- I Bellezza
- Dipartimento di Medicina Sperimentale, Università di Perugia, Polo Unico Sant'Andrea delle Fratte, p.le Gambuli, Perugia, 06132; Italia
| | - L Gatticchi
- Dipartimento di Medicina Sperimentale, Università di Perugia, Polo Unico Sant'Andrea delle Fratte, p.le Gambuli, Perugia, 06132; Italia
| | - R del Sordo
- Dipartimento di Medicina Sperimentale, Università di Perugia, Polo Unico Sant'Andrea delle Fratte, p.le Gambuli, Perugia, 06132; Italia
| | - M J Peirce
- Dipartimento di Medicina Sperimentale, Università di Perugia, Polo Unico Sant'Andrea delle Fratte, p.le Gambuli, Perugia, 06132; Italia
| | - A Sidoni
- Dipartimento di Medicina Sperimentale, Università di Perugia, Polo Unico Sant'Andrea delle Fratte, p.le Gambuli, Perugia, 06132; Italia
| | - R Roberti
- Dipartimento di Medicina Sperimentale, Università di Perugia, Polo Unico Sant'Andrea delle Fratte, p.le Gambuli, Perugia, 06132; Italia
| | - A Minelli
- Dipartimento di Medicina Sperimentale, Università di Perugia, Polo Unico Sant'Andrea delle Fratte, p.le Gambuli, Perugia, 06132; Italia
| |
Collapse
|
42
|
Sánchez-Guijo A, Oji V, Hartmann MF, Schuppe HC, Traupe H, Wudy SA. High levels of oxysterol sulfates in serum of patients with steroid sulfatase deficiency. J Lipid Res 2014; 56:403-12. [PMID: 25502769 DOI: 10.1194/jlr.m055608] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Steroid sulfatase (STS) deficiency is the underlying cause of the skin condition known as recessive X-linked ichthyosis (RXLI). RXLI patients show scales on their skin caused by high concentrations of cholesterol sulfate (CS), as they are not capable of releasing the sulfate group from its structure to obtain free cholesterol. CS has been reported, so far, as the sole sulfated steroid with increased concentrations in the blood of RXLI patients. A non-targeted LC-MS approach in negative mode detection (LC-MS precursor ion scan mode) was applied to serum samples of 12 RXLI patients and 19 healthy males. We found that CS was not the only sulfated compound consistently elevated in RXLI patients, because a group of compounds with a m/z of 481 was found in high concentrations too. Further LC-MS/MS demonstrated that the main contributor to the m/z 481 signal in RXLI serum is 27-hydroxycholesterol-3-sulfate (27OHC3S). Accordingly, a new method for 27OHC3S quantification in the context of RXLI has been developed and validated. Other hydroxycholesterol sulfate compounds were elevated as well in RXLI patients.
Collapse
Affiliation(s)
- Alberto Sánchez-Guijo
- Steroid Research and Mass Spectrometry Unit, Division of Pediatric Endocrinology and Diabetology, Center of Child and Adolescent Medicine, Justus-Liebig University, 35392 Giessen, Germany
| | - Vinzenz Oji
- Department of Dermatology, University of Münster, 48149 Münster, Germany
| | - Michaela F Hartmann
- Steroid Research and Mass Spectrometry Unit, Division of Pediatric Endocrinology and Diabetology, Center of Child and Adolescent Medicine, Justus-Liebig University, 35392 Giessen, Germany
| | - Hans-Christian Schuppe
- Clinic of Urology, Pediatric Urology and Andrology, Justus-Liebig-University, 35385 Giessen, Germany
| | - Heiko Traupe
- Department of Dermatology, University of Münster, 48149 Münster, Germany
| | - Stefan A Wudy
- Steroid Research and Mass Spectrometry Unit, Division of Pediatric Endocrinology and Diabetology, Center of Child and Adolescent Medicine, Justus-Liebig University, 35392 Giessen, Germany
| |
Collapse
|
43
|
The Influence of an Obesogenic Diet on Oxysterol Metabolism in C57BL/6J Mice. CHOLESTEROL 2014; 2014:843468. [PMID: 24672716 PMCID: PMC3941159 DOI: 10.1155/2014/843468] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 12/18/2013] [Accepted: 12/21/2013] [Indexed: 11/18/2022]
Abstract
Our current understanding of oxysterol metabolism during different disease states such as obesity and dyslipidemia is limited. Therefore, the aim of this study was to determine the effect of diet-induced obesity on the tissue distribution of various oxysterols and the mRNA expression of key enzymes involved in oxysterol metabolism. To induce obesity, male C57BL/6J mice were fed a high fat-cholesterol diet for 24 weeks. Following diet-induced obesity, plasma levels of 4β-hydroxycholesterol, 5,6α-epoxycholesterol, 5,6β-epoxycholesterol, 7α-hydroxycholesterol, 7β-hydroxycholesterol, and 27-hydroxycholesterol were significantly (P < 0.05) increased. In the liver and adipose tissue of the obese mice, 4β-hydroxycholesterol was significantly (P < 0.05) increased, whereas 27-hydroxycholesterol was increased only in the adipose tissue. No significant changes in either hepatic or adipose tissue mRNA expression were observed for oxysterol synthesizing enzymes 4β-hydroxylase, 27-hydroxylase, or 7α-hydroxylase. Hepatic mRNA expression of SULT2B1b, a key enzyme involved in oxysterol detoxification, was significantly (P < 0.05) elevated in the obese mice. Interestingly, the appearance of the large HDL1 lipoprotein was observed with increased oxysterol synthesis during obesity. In diet-induced obese mice, dietary intake and endogenous enzymatic synthesis of oxysterols could not account for the increased oxysterol levels, suggesting that nonenzymatic cholesterol oxidation pathways may be responsible for the changes in oxysterol metabolism.
Collapse
|
44
|
Shi X, Cheng Q, Xu L, Yan J, Jiang M, He J, Xu M, Stefanovic-Racic M, Sipula I, O'Doherty RM, Ren S, Xie W. Cholesterol sulfate and cholesterol sulfotransferase inhibit gluconeogenesis by targeting hepatocyte nuclear factor 4α. Mol Cell Biol 2014; 34:485-97. [PMID: 24277929 PMCID: PMC3911511 DOI: 10.1128/mcb.01094-13] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 09/20/2013] [Accepted: 11/11/2013] [Indexed: 11/20/2022] Open
Abstract
Sulfotransferase (SULT)-mediated sulfation represents a critical mechanism in regulating the chemical and functional homeostasis of endogenous and exogenous molecules. The cholesterol sulfotransferase SULT2B1b catalyzes the sulfoconjugation of cholesterol to synthesize cholesterol sulfate (CS). In this study, we showed that the expression of SULT2B1b in the liver was induced in obese mice and during the transition from the fasted to the fed state, suggesting that the regulation of SULT2B1b is physiologically relevant. CS and SULT2B1b inhibited gluconeogenesis by targeting the gluconeogenic factor hepatocyte nuclear factor 4α (HNF4α) in both cell cultures and transgenic mice. Treatment of mice with CS or transgenic overexpression of the CS-generating enzyme SULT2B1b in the liver inhibited hepatic gluconeogenesis and alleviated metabolic abnormalities both in mice with diet-induced obesity (DIO) and in leptin-deficient (ob/ob) mice. Mechanistically, CS and SULT2B1b inhibited gluconeogenesis by suppressing the expression of acetyl coenzyme A (acetyl-CoA) synthetase (Acss), leading to decreased acetylation and nuclear exclusion of HNF4α. Our results also suggested that leptin is a potential effector of SULT2B1b in improving metabolic function. We conclude that SULT2B1b and its enzymatic by-product CS are important metabolic regulators that control glucose metabolism, suggesting CS as a potential therapeutic agent and SULT2B1b as a potential therapeutic target for metabolic disorders.
Collapse
MESH Headings
- Acetylation/drug effects
- Animals
- Blotting, Western
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cells, Cultured
- Cholesterol Esters/metabolism
- Cholesterol Esters/pharmacology
- Coenzyme A Ligases/genetics
- Coenzyme A Ligases/metabolism
- Colforsin/pharmacology
- Diet, High-Fat/adverse effects
- Gene Expression/drug effects
- Gluconeogenesis/drug effects
- Gluconeogenesis/genetics
- Glucose/metabolism
- Hepatocyte Nuclear Factor 4/genetics
- Hepatocyte Nuclear Factor 4/metabolism
- Humans
- Insulin Resistance
- Mice
- Mice, Inbred C57BL
- Mice, Obese
- Mice, Transgenic
- Obesity/etiology
- Obesity/genetics
- Obesity/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sulfotransferases/genetics
- Sulfotransferases/metabolism
Collapse
Affiliation(s)
- Xiongjie Shi
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Qiuqiong Cheng
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Leyuan Xu
- Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia, USA
| | - Jiong Yan
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Mengxi Jiang
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Jinhan He
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Meishu Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Maja Stefanovic-Racic
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ian Sipula
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert Martin O'Doherty
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shunlin Ren
- Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia, USA
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
45
|
Kodama S, Negishi M. Sulfotransferase genes: regulation by nuclear receptors in response to xeno/endo-biotics. Drug Metab Rev 2013; 45:441-9. [PMID: 24025090 DOI: 10.3109/03602532.2013.835630] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Pregnane X receptor (PXR) and constitutive active/androstane receptor (CAR), members of the nuclear receptor superfamily, are two major xeno-sensing transcription factors. They can be activated by a broad range of lipophilic xenobiotics including therapeutics drugs. In addition to xenobiotics, endogenous compounds such as steroid hormones and bile acids can also activate PXR and/or CAR. These nuclear receptors regulate genes that encode enzymes and transporters that metabolize and excrete both xenobiotics and endobiotics. Sulfotransferases (SULTs) are a group of these enzymes and sulfate xenobiotics for detoxification. In general, inactivation by sulfation constitutes the mechanism to maintain homeostasis of endobiotics. Thus, deciphering the molecular mechanism by which PXR and CAR regulate SULT genes is critical for understanding the roles of SULTs in the alterations of physiological and pathophysiological processes caused by drug treatment or environmental exposures.
Collapse
Affiliation(s)
- Susumu Kodama
- Division of Drug Metabolism and Molecular Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University , Sendai , Japan and
| | | |
Collapse
|
46
|
Falany CN, Rohn-Glowacki KJ. SULT2B1: unique properties and characteristics of a hydroxysteroid sulfotransferase family. Drug Metab Rev 2013; 45:388-400. [PMID: 24020383 DOI: 10.3109/03602532.2013.835609] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The SULT2b gene family consists of a single gene capable of generating two functional transcripts utilizing different transcriptional start sites in the first exon. This results in the translation of two separate proteins, SULT2B1a and SULT2B1b, with different amino-terminal peptides and approximately 95% identical sequences. The second distinguishing feature of the SULT2B isoforms is the proline/serine-rich carboxy-terminal sequence. To date, presence of the SULT2B gene appears limited to mammals and there is also only limited conservation of structure or sequence of the carboxy-terminal peptide. Although both SULT2B1 messages are present in human tissues, to date, only the SULT2B1b protein has been detected in the tissues investigated. In contrast, selective expression of SULT2B1a has been detected in rodent brain, whereas SULT2B1b was expressed in skin and intestine. Characterization of the SULT2B1 isoforms has been limited by the inability to isolate reliably active SULT2B1b from tissues or cells. SULT2B1 cDNAs can be expressed in Escherichia coli and the expressed active enzymes show selectivity for sulfation of 3β-hydroxysteroids. SULT2B1b due to the binding properties of the amino-terminal peptides also shows high cholesterol sulfation activity. Although human SULT2B1b displays significant substrate cross-reactivity with SULT2A1, the isoforms have different tissue expression patterns. Human SULT2B1b also shows nuclear localization in selected tissues that appears related to serine phosphorylation of the carboxy-terminal peptide. Overall, the understanding of the properties and function of the SULT2B1 isoforms is limited and the structural variability of the unique amino- and carboxy-sequences suggests significant species differences that need to be investigated.
Collapse
Affiliation(s)
- Charles N Falany
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham , Birmingham, AL , USA
| | | |
Collapse
|
47
|
Acimovic J, Lövgren-Sandblom A, Olin M, Ali Z, Heverin M, Schüle R, Schöls L, Fischler B, Fickert P, Trauner M, Björkhem I. Sulphatation does not appear to be a protective mechanism to prevent oxysterol accumulation in humans and mice. PLoS One 2013; 8:e68031. [PMID: 23844150 PMCID: PMC3700920 DOI: 10.1371/journal.pone.0068031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 05/24/2013] [Indexed: 01/10/2023] Open
Abstract
24S- and 27-hydroxycholesterol (24OHC and 27OHC) are potent regulators of different biochemical systems in vitro and are the major circulating oxysterols. A small fraction of these oxysterols has been reported to be sulphated but there are no detailed studies. We considered the possibility that sulphatation is a protective mechanism preventing accumulation of free oxysterols. Using an accurate assay we found the sulphated fraction of 24OHC and 27OHC in circulation of adults to be less than 15% of total. In two patients with a mutation in CYP7B1 and markedly increased levels of 27OHC the sulphated fraction was 8% and 10% respectively. Infants with severe neonatal cholestasis had however markedly increased sulphate fraction of the above oxysterols. In untreated mice the degree of sulphatation of 24OHC and 27OHC in serum varied between 0 and 16%. Similar degree of sulphatation was found in two mouse models with markedly increased levels of 27OHC and 24OHC respectively. Bile duct ligated mice had higher levels of oxysterols than sham-operated controls but the sulphate fraction was not increased. We conclude that a primary increase in the levels of the oxysterols due to increased synthesis or reduced metabolism in adults and mice does not induce increased sulphatation.
Collapse
Affiliation(s)
- Jure Acimovic
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ikegawa S. [Detailed characterization of bile acid and glucocorticoid world by mass spectrometry]. YAKUGAKU ZASSHI 2013; 133:661-79. [PMID: 23728093 DOI: 10.1248/yakushi.13-00099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Nobel Prize in Chemistry for 2002 was shared by John B. Fenn and Koichi Tanaka "for their development of soft desorption methods for mass spectrometric analyses of biological macromolecules". Indeed, electrospray ionization and soft laser desorption ionization have proved to be of great value in "omics", such as metabolomics, transcriptomics and proteomics in providing a systematic and quantitative approach to the study of biological systems and networks. Moreover, these techniques have made great contributions to metabolic studies that are used for development of new drugs, as well as to the diagnosis of diseases including cancer based on the specific and sensitive detection of molecular biomarkers. In this article, we describe our recent results on characterization of bile acid metabolism in hepatobiliary disease as well as measurement of conjugated urinary tetrahydrocorticosteroids for assessment of altered corticoid metabolism in endocrine disease and the metabolic syndrome.
Collapse
Affiliation(s)
- Shigeo Ikegawa
- Faculty of Pharmaceutical Sciences, Kinki University, 3-4-1 Kowakae, Higashi-osaka, Osaka 577-8502, Japan.
| |
Collapse
|
49
|
Xu L, Kim JK, Bai Q, Zhang X, Kakiyama G, Min HK, Sanyal AJ, Pandak WM, Ren S. 5-cholesten-3β,25-diol 3-sulfate decreases lipid accumulation in diet-induced nonalcoholic fatty liver disease mouse model. Mol Pharmacol 2013; 83:648-58. [PMID: 23258548 PMCID: PMC3583496 DOI: 10.1124/mol.112.081505] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2012] [Accepted: 12/20/2012] [Indexed: 12/19/2022] Open
Abstract
Sterol regulatory element-binding protein-1c (SREBP-1c) increases lipogenesis at the transcriptional level, and its expression is upregulated by liver X receptor α (LXRα). The LXRα/SREBP-1c signaling may play a crucial role in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). We previously reported that a cholesterol metabolite, 5-cholesten-3β,25-diol 3-sulfate (25HC3S), inhibits the LXRα signaling and reduces lipogenesis by decreasing SREBP-1c expression in primary hepatocytes. The present study aims to investigate the effects of 25HC3S on lipid homeostasis in diet-induced NAFLD mouse models. NAFLD was induced by feeding a high-fat diet (HFD) in C57BL/6J mice. The effects of 25HC3S on lipid homeostasis, inflammatory responses, and insulin sensitivity were evaluated after acute treatments or long-term treatments. Acute treatments with 25HC3S decreased serum lipid levels, and long-term treatments decreased hepatic lipid accumulation in the NAFLD mice. Gene expression analysis showed that 25HC3S significantly suppressed the SREBP-1c signaling pathway that was associated with the suppression of the key enzymes involved in lipogenesis: fatty acid synthase, acetyl-CoA carboxylase 1, and glycerol-3-phosphate acyltransferase. In addition, 25HC3S significantly reduced HFD-induced hepatic inflammation as evidenced by decreasing tumor necrosis factor and interleukin 1 α/β mRNA levels. A glucose tolerance test and insulin tolerance test showed that 25HC3S administration improved HFD-induced insulin resistance. The present results indicate that 25HC3S as a potent endogenous regulator decreases lipogenesis, and oxysterol sulfation can be a key protective regulatory pathway against lipid accumulation and lipid-induced inflammation in vivo.
Collapse
Affiliation(s)
- Leyuan Xu
- McGuire Veterans Affairs Medical Center/Virginia Commonwealth University, Research 151, Richmond, VA 23249, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Zhang X, Bai Q, Xu L, Kakiyama G, Pandak WM, Zhang Z, Ren S. Cytosolic sulfotransferase 2B1b promotes hepatocyte proliferation gene expression in vivo and in vitro. Am J Physiol Gastrointest Liver Physiol 2012; 303:G344-55. [PMID: 22679001 PMCID: PMC3423104 DOI: 10.1152/ajpgi.00403.2011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cytosolic sulfotransferase 2B1b (SULT2B1b) catalyzes the sulfation of 3β-hydroxysteroids and functions as a selective cholesterol and oxysterol sulfotransferase. Activation of liver X receptors (LXRs) by oxysterols has been known to be an antiproliferative factor. Overexpression of SULT2B1b impairs LXR's response to oxysterols, by which it regulates lipid metabolism. The aim of this study was to investigate in vivo and in vitro effects of SULT2B1b on liver proliferation and the underlying mechanisms. Primary rat hepatocytes and C57BL/6 mice were infected with adenovirus encoding SULT2B1b. Liver proliferation was determined by measuring the proliferating cell nuclear antigen (PCNA) immunostaining labeling index. The correlation between SULT2B1b and PCNA expression in mouse liver tissues was determined by double immunofluorescence. Gene expressions were evaluated by quantitative real-time PCR and Western blot analysis. SULT2B1b overexpression in mouse liver tissues increased PCNA-positive cells in a dose- and time-dependent manner. The increased expression of PCNA in mouse liver tissues was only observed in the SULT2B1b transgenic cells. Small interference RNA SULT2B1b significantly inhibited cell cycle regulatory gene expressions in primary rat hepatocytes. LXR activation by T0901317 effectively suppressed SULT2B1b-induced gene expression in vivo and in vitro. SULT2B1b may promote hepatocyte proliferation by inactivating oxysterol/LXR signaling.
Collapse
Affiliation(s)
- Xin Zhang
- 1Department of Pathology, Fudan University Shanghai Medical College, Shanghai, China; and ,2Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia
| | - Qianming Bai
- 1Department of Pathology, Fudan University Shanghai Medical College, Shanghai, China; and ,2Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia
| | - Leyuan Xu
- 2Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia
| | - Genta Kakiyama
- 2Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia
| | - William M. Pandak
- 2Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia
| | - Zhigang Zhang
- 1Department of Pathology, Fudan University Shanghai Medical College, Shanghai, China; and
| | - Shunlin Ren
- 2Department of Medicine, Virginia Commonwealth University/Veterans Affairs McGuire Medical Center, Richmond, Virginia
| |
Collapse
|