1
|
Yamauchi Y, Abe-Dohmae S, Iwamoto N, Sato R, Yokoyama S. ABCA1 deficiency causes tissue-specific dysregulation of the SREBP2 pathway in mice. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159546. [PMID: 39089642 DOI: 10.1016/j.bbalip.2024.159546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
ABCA1 plays an essential role in the formation of high-density lipoprotein (HDL), and its mutations cause Tangier disease (TD), a familial HDL deficiency. In addition to the disappearance of HDL, TD patients exhibit cholesterol deposition in peripheral tissues through a mechanism poorly understood, which may contribute to the development of premature atherosclerosis. We and others previously showed that ABCA1 deficiency causes hyperactivation of the SREBP2 pathway in vitro. Here, we show using Abca1 knockout mice that ABCA1 deficiency leads to tissue-specific dysregulation of SREBP2 activity in a nutritional status-dependent manner, which may underlie the pathophysiology of TD.
Collapse
Affiliation(s)
- Yoshio Yamauchi
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan; Department of Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Sumiko Abe-Dohmae
- Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan; Department of Food and Nutritional Sciences, Chubu University, Kasugai 487-8501, Japan
| | - Noriyuki Iwamoto
- Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Ryuichiro Sato
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Shinji Yokoyama
- Department of Food and Nutritional Sciences, Chubu University, Kasugai 487-8501, Japan
| |
Collapse
|
2
|
Yu L, Long Q, Zhang Y, Liu Y, Guo Z, Cao X, Qin F, Xu Y, Qian Q, Gao B, Chen J, Liu J, Zeng Y, Teng Z. Bidirectional Mendelian randomization analysis of plasma lipidome and psychiatric disorders. J Affect Disord 2024:S0165-0327(24)01757-9. [PMID: 39442703 DOI: 10.1016/j.jad.2024.10.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Evidence from observational studies and clinical experiments suggests a close association between plasma lipidome and psychiatric disorders. However, the causal relationship between plasma lipidome and psychiatric disorders remains insufficiently determined. Plasma lipidome are relatively easy to measure and regulate clinically, and they play a crucial role in neuronal signal transduction, making them a focus of interest as potential therapeutic targets for psychiatric disorders. METHODS In this study, we utilized the latest Finnish population-based genome-wide association study (GWAS) data on 179 lipid species. We downloaded data on five psychiatric disorders from the IEU database, including schizophrenia, bipolar disorder, depression, autism from the PGC consortium, and anxiety disorder from the Neale lab. Using two-sample bidirectional Mendelian randomization (MR) analysis, we assessed the relationship between these 179 lipid species and the risk of the five psychiatric disorders. To validate the assumptions of Mendelian randomization, we conducted tests for horizontal pleiotropy and heterogeneity. RESULTS After applying FDR correction to assess the relationship between 179 lipid species traits and the risk of five psychiatric disorders, our analysis provided evidence of a causal relationship specifically between genetic susceptibility in the plasma lipidome and bipolar disorder. This relationship notably involves eight phosphatidylcholines (PCs) and two sterols, with PCs displaying a dual and complex role in the disorder. Reverse Mendelian randomization (MR) analysis did not reveal a significant causal impact of psychiatric disorders on the plasma lipidome. LIMITATIONS Despite using two-sample bidirectional Mendelian randomization analysis, the complex biological pathways and potential confounding factors may still affect the accuracy of the causal relationships. The impact of genetic variations on the lipidome and psychiatric disorders may involve multiple mechanisms, which cannot be fully elucidated in this study. CONCLUSION This study identified a causal relationship between genetic susceptibility in plasma lipidome and bipolar disorder, indicating that plasma lipidome may influence the risk of psychiatric disorders and providing direction for exploring them as potential intervention targets. The findings not only deepen our understanding of the etiology of psychiatric disorders but also provide a critical theoretical foundation for future clinical interventions and prevention strategies, potentially contributing to the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Ling Yu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qing Long
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunqiao Zhang
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China; Department of Psychiatry, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Yilin Liu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ziyi Guo
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiang Cao
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fuyi Qin
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yangyang Xu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qingqing Qian
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Biyao Gao
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jian Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Jie Liu
- Department of Dermatology and Venereology, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Yong Zeng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China; Department of Psychiatry, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China.
| | - Zhaowei Teng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
3
|
Karabekmez ME. Insights into yeast response to chemotherapeutic agent through time series genome-scale metabolic models. Biotechnol Bioeng 2024; 121:3351-3359. [PMID: 39199017 DOI: 10.1002/bit.28833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/17/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Organism-specific genome-scale metabolic models (GSMMs) can unveil molecular mechanisms within cells and are commonly used in diverse applications, from synthetic biology, biotechnology, and systems biology to metabolic engineering. There are limited studies incorporating time-series transcriptomics in GSMM simulations. Yeast is an easy-to-manipulate model organism for tumor research. Here, a novel approach (TS-GSMM) was proposed to integrate time-series transcriptomics with GSMMs to narrow down the feasible solution space of all possible flux distributions and attain time-series flux samples. The flux samples were clustered using machine learning techniques, and the clusters' functional analysis was performed using reaction set enrichment analysis. A time series transcriptomics response of Yeast cells to a chemotherapeutic reagent-doxorubicin-was mapped onto a Yeast GSMM. Eleven flux clusters were obtained with our approach, and pathway dynamics were displayed. Induction of fluxes related to bicarbonate formation and transport, ergosterol and spermidine transport, and ATP production were captured. Integrating time-series transcriptomics data with GSMMs is a promising approach to reveal pathway dynamics without any kinetic modeling and detects pathways that cannot be identified through transcriptomics-only analysis. The codes are available at https://github.com/karabekmez/TS-GSMM.
Collapse
|
4
|
Yan P, Zhao D. Association between serum total cholesterol and the development of gastric cancer: A two-way two-sample Mendelian randomization study. Medicine (Baltimore) 2024; 103:e38900. [PMID: 38996131 PMCID: PMC11245271 DOI: 10.1097/md.0000000000038900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Previous epidemiologic studies have suggested a potential negative correlation between total cholesterol (TC) and Gastric cancer (GC); however, several observational studies have shown conflicting results and have failed to provide definitive evidence for a causal relationship between TC and GC. Therefore, we conducted a 2-sample bidirectional Mendelian randomization (MR) study to explore the genetic correlation and potential causal relationship between the 2 variables. We screened for single nucleotide polymorphisms (SNPs) associated with TC and GC utilizing a large-scale genome-wide association study (GWAS) public database. The causal relationship was analyzed using 5 MR analysis methods: inverse variance weighting (IVW), weighted median, MR-Egger regression, weighted mode, and simple mode. Additionally, reverse MR analysis was performed to evaluate the possibility of reverse causality. Sensitivity analyses were conducted, including heterogeneity tests, horizontal multiple validity tests, and leave-one-out tests. After meticulous screening, 79 SNPs were identified as instrumental variables (IVs). The IVW method revealed a causal relationship between TC and GC (OR = 0.844; 95% CI: 0.741-0.961; P = .01). Sensitivity analyses did not detect significant horizontal pleiotropy. Though heterogeneity was observed in the forward MR analysis (IVW, Qp = 0.0006), the results remained reliable as we utilized the IVW random-effects model as the primary analytical method. Furthermore, inverse MR analysis found no evidence of reverse causality between TC and GC, effectively ruling out the influence of GC on the reverse causality of TC. Our MR study provided evidence of a causal association between TC and GC, suggesting that TC acts as a protective factor against GC due to its negative association with the disease.
Collapse
Affiliation(s)
- Peng Yan
- Department of Medical Oncology, Lixin County People’s Hospital, Bozhou, Anhui, China
| | - Dong Zhao
- Department of Medical Oncology, Lixin County People’s Hospital, Bozhou, Anhui, China
| |
Collapse
|
5
|
Zhou H, Ren S, Yang Y, Qin Y, Guo T, Zhou Y, Zhang Y, Ma L. Transgenerational toxicity induced by maternal AFB 1 exposure in Caenorhabditis elegans associated with underlying epigenetic regulations. Food Chem Toxicol 2024; 187:114599. [PMID: 38490352 DOI: 10.1016/j.fct.2024.114599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
Aflatoxin B1 (AFB1), usually seriously contaminates in grain and oil foods or feed, displayed significant acute and chronic toxic effects in human and animal populations. However, little is known about the transgenerational toxic effects induced by a maternal AFB1 intake at a lower dose on offspring. In our study, only parental wild-type Caenorhabditis elegans was exposed to AFB1 (0-8 μg/ml) and the following three filial generations were grown on AFB1-free NGM. Results showed that the toxic effects of AFB1 on the growth (body length) and reproduction (brood size, generation time and morphology of gonad arm) can be transmitted through generations. Moreover, the levels of MMP and ATP were irreversibly inhibited in the filial generations. By using RNomics and molecular biology techniques, we found that steroid biosynthesis, phagosome, valine/leucine/isoleucine biosynthesis and oxidative phosphorylation (p < 0.05) were the core signaling pathways to exert the transgenerational toxic effects on nematodes. Also, notably increased histone methylation level at H3K36me3 was observed in the first generation. Taken together, our study demonstrated that AFB1 has notable transgenerational toxic effects, which were resulted from the complex regulatory network of various miRNAs, mRNAs and epigenetic modification in C. elegans.
Collapse
Affiliation(s)
- Hongyuan Zhou
- College of Food Science, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing, 400715, China; Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, China.
| | - Sirui Ren
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Yulian Yang
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Yuxian Qin
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Ting Guo
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Ying Zhou
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Yuhao Zhang
- College of Food Science, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing, 400715, China; Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Chongqing, 400715, China
| | - Liang Ma
- College of Food Science, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing, 400715, China; Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, China; Key Laboratory of Condiment Supervision Technology for State Market Regulation, Chongqing, 401121, China.
| |
Collapse
|
6
|
Decker NS, Johnson T, Le Cornet C, Behrens S, Obi N, Kaaks R, Chang-Claude J, Fortner RT. Associations between lifestyle, health, and clinical characteristics and circulating oxysterols and cholesterol precursors in women diagnosed with breast cancer: a cross-sectional study. Sci Rep 2024; 14:4977. [PMID: 38424253 PMCID: PMC10904394 DOI: 10.1038/s41598-024-55316-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/22/2024] [Indexed: 03/02/2024] Open
Abstract
Despite increasing evidence that cholesterol precursors and oxysterols, oxidized cholesterol metabolites, play a role in numerous pathological processes and diseases including breast cancer, little is known about correlates of these sterols in women with breast cancer. In this study, 2282 women with breast cancer and blood draw post diagnosis were included and cross-sectional associations between circulating levels of 15 sterols/oxysterols and (a) lifestyle, anthropometric, reproductive characteristics, (b) comorbidities and medication use, and (c) breast cancer tumor and treatment characteristics were calculated using generalized linear models. Obesity was strongly associated with circulating levels of 7-dehydrocholesterol (DC) (body mass index ≥ 30 vs. 18.5-24.9 kg/m2: 51.7% difference) and 7-ketocholesterol (KC) (40.0% difference). After adjustment for BMI, comorbidities such as cardiovascular disease were associated with higher levels of 7-DC (26.1% difference) and lower levels of desmosterol (- 16.4% difference). Breast cancer tumor characteristics including hormone receptor status, tumor stage, and endocrine therapy were associated with lanosterol, 24-DHLan, 7b-HC, and THC (e.g., THC; tumor stage IIIa vs. I: 36.9% difference). Weaker associations were observed for lifestyle characteristics and for any of the other oxysterols. The findings of this study suggest that cholesterol precursors are strongly associated with metabolic factors, while oxysterols are associated with breast cancer tumor characteristics, warranting further investigation into the role of cholesterol precursors and oxysterols in women with breast cancer and other populations.
Collapse
Affiliation(s)
- Nina Sophia Decker
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Theron Johnson
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Charlotte Le Cornet
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine Behrens
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadia Obi
- Institute for Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute for Occupational and Maritime Medicine Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- University Cancer Center Hamburg, Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Renée Turzanski Fortner
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Research, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway.
| |
Collapse
|
7
|
Saito H, Nishimura M, Sato R, Yamauchi Y. Quantitative Determination of Cholesterol Hydroxylase Specificities by GC-MS/MS in Living Mammalian Cells. Bio Protoc 2024; 14:e4924. [PMID: 38268974 PMCID: PMC10804311 DOI: 10.21769/bioprotoc.4924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 12/07/2023] [Accepted: 12/13/2023] [Indexed: 01/26/2024] Open
Abstract
Cholesterol is oxygenated by a variety of cholesterol hydroxylases; oxysterols play diverse important roles in physiological and pathophysiological conditions by regulating several transcription factors and cell-surface receptors. Each oxysterol has distinct and overlapping functions. The expression of cholesterol hydroxylases is highly regulated, but their physiological and pathophysiological roles are not fully understood. Although the activity of cholesterol hydroxylases has been characterized biochemically using radiolabeled cholesterol as the substrate, their specificities remain to be comprehensively determined quantitatively. To better understand their roles, a highly sensitive method to measure the amount of various oxysterols synthesized by cholesterol hydroxylases in living mammalian cells is required. Our method described here, with gas chromatography coupled with tandem mass spectrometry (GC-MS/MS), can quantitatively determine a series of oxysterols endogenously synthesized by forced expression of one of the four major cholesterol hydroxylases-CH25H, CYP7A1, CYP27A1, and CYP46A1-or induction of CH25H expression by a physiological stimulus. This protocol can also simultaneously measure the amount of intermediate sterols, which serve as markers for cellular cholesterol synthesis activity. Key features • Allows measuring the amount of a variety of oxysterols synthesized endogenously by cholesterol hydroxylases using GC-MS/MS. • Comprehensive and quantitative analysis of cholesterol hydroxylase specificities in living mammalian cells. • Simultaneous quantification of intermediate sterols to assess cholesterol synthesis activity.
Collapse
Affiliation(s)
- Hodaka Saito
- Laboratory of Food Biochemistry, Department of
Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences,
The University of Tokyo, Tokyo, Japan
| | - Mizuki Nishimura
- Laboratory of Food Biochemistry, Department of
Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences,
The University of Tokyo, Tokyo, Japan
| | - Ryuichiro Sato
- Nutri-Life Science Laboratory, Department of Applied
Biological Chemistry, Graduate School of Agricultural and Life Sciences, The
University of Tokyo, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and
Development, Tokyo, Japan
| | - Yoshio Yamauchi
- Laboratory of Food Biochemistry, Department of
Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences,
The University of Tokyo, Tokyo, Japan
- Nutri-Life Science Laboratory, Department of Applied
Biological Chemistry, Graduate School of Agricultural and Life Sciences, The
University of Tokyo, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and
Development, Tokyo, Japan
| |
Collapse
|
8
|
Bell CF, Lei X, Haas A, Baylis RA, Gao H, Luo L, Giordano SH, Wehner MR, Nead KT, Leeper NJ. Risk of Cancer After Diagnosis of Cardiovascular Disease. JACC CardioOncol 2023; 5:431-440. [PMID: 37614573 PMCID: PMC10443115 DOI: 10.1016/j.jaccao.2023.01.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 08/25/2023] Open
Abstract
Background Cardiovascular disease (CVD) and cancer share several risk factors. Although preclinical models show that various types of CVD can accelerate cancer progression, clinical studies have not determined the impact of atherosclerosis on cancer risk. Objectives The objective of this study was to determine whether CVD, especially atherosclerotic CVD, is independently associated with incident cancer. Methods Using IBM MarketScan claims data from over 130 million individuals, 27 million cancer-free subjects with a minimum of 36 months of follow-up data were identified. Individuals were stratified by presence or absence of CVD, time-varying analysis with multivariable adjustment for cardiovascular risk factors was performed, and cumulative risk of cancer was calculated. Additional analyses were performed according to CVD type (atherosclerotic vs nonatherosclerotic) and cancer subtype. Results Among 27,195,088 individuals, those with CVD were 13% more likely to develop cancer than those without CVD (HR: 1.13; 95% CI: 1.12-1.13). Results were more pronounced for individuals with atherosclerotic CVD (aCVD), who had a higher risk of cancer than those without CVD (HR: 1.20; 95% CI: 1.19-1.21). aCVD also conferred a higher risk of cancer compared with those with nonatherosclerotic CVD (HR: 1.11; 95% CI: 1.11-1.12). Cancer subtype analyses showed specific associations of aCVD with several malignancies, including lung, bladder, liver, colon, and other hematologic cancers. Conclusions Individuals with CVD have an increased risk of developing cancer compared with those without CVD. This association may be driven in part by the relationship of atherosclerosis with specific cancer subtypes, which persists after controlling for conventional risk factors.
Collapse
Affiliation(s)
- Caitlin F. Bell
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, USA
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, USA
- Stanford Cardiovascular Institute, Stanford, California, USA
| | - Xiudong Lei
- Department of Health Services Research, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Allen Haas
- Department of Health Services Research, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Richard A. Baylis
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, USA
- Stanford Cardiovascular Institute, Stanford, California, USA
| | - Hua Gao
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, USA
- Stanford Cardiovascular Institute, Stanford, California, USA
| | - Lingfeng Luo
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, USA
- Stanford Cardiovascular Institute, Stanford, California, USA
| | - Sharon H. Giordano
- Department of Health Services Research, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mackenzie R. Wehner
- Department of Health Services Research, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Dermatology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kevin T. Nead
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nicholas J. Leeper
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, USA
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, USA
- Stanford Cardiovascular Institute, Stanford, California, USA
| |
Collapse
|
9
|
Tkemaladze T, Bratland E, Bregvadze K, Shatirishvili T, Tatishvili N, Abzianidze E, Houge G, Douzgou S. MSMO1 deficiency: a potentially partially treatable, ultrarare neurodevelopmental disorder with psoriasiform dermatitis, alopecia and polydactyly. Clin Dysmorphol 2023; 32:97-105. [PMID: 37195326 DOI: 10.1097/mcd.0000000000000461] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
MSMO1 deficiency (OMIM #616834) is an ultrarare autosomal recessive disorder of distal cholesterol metabolism with only five cases reported to date. The disorder is caused by missense variants in the MSMO1 gene encoding methylsterol monooxygenase 1, leading to the accumulation of methylsterols. Clinically, MSMO1 deficiency is characterized by growth and developmental delay, often in association with congenital cataracts, microcephaly, psoriasiform dermatitis and immune dysfunction. Treatment with oral and topical cholesterol supplements and statins was reported to improve the biochemical, immunological, and cutaneous findings, supporting a potential treatment following the precision diagnosis of MSMO1 deficiency. We describe two siblings from a consanguineous family presenting with novel clinical features of polydactyly, alopecia and spasticity. Whole-exome sequencing revealed a novel, homozygous c.548A > C, p.(Glu183Ala) variant. Based on previously published treatment algorithms, we initiated a modified dosage regime with systemic cholesterol supplementation, statins and bile acid along with topical application of a cholesterol/statin formulation. This resulted in a marked improvement of psoriasiform dermatitis and some hair growth.
Collapse
Affiliation(s)
- Tinatin Tkemaladze
- Department of Molecular and Medical Genetics, Tbilisi State Medical University
- Department of Child Neurology, M. Iashvili Children's Central Hospital, Tbilisi, Georgia
| | - Eirik Bratland
- Department of Medical Genetics, Haukeland University Hospital
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kakha Bregvadze
- Department of Molecular and Medical Genetics, Tbilisi State Medical University
| | - Teona Shatirishvili
- Department of Child Neurology, M. Iashvili Children's Central Hospital, Tbilisi, Georgia
| | - Nino Tatishvili
- Department of Child Neurology, M. Iashvili Children's Central Hospital, Tbilisi, Georgia
| | - Elene Abzianidze
- Department of Molecular and Medical Genetics, Tbilisi State Medical University
| | - Gunnar Houge
- Department of Medical Genetics, Haukeland University Hospital
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Sofia Douzgou
- Department of Medical Genetics, Haukeland University Hospital
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
10
|
Shi M, Zheng C, Sun Y, Li X, He G, Cao J, Tan B, Xie S. Effects of Dietary Chenodeoxycholic Acid Supplementation in a Low Fishmeal Diet Containing Clostridium autoethanogenum Protein on Growth, Lipid and Cholesterol Metabolism, and Hepatopancreas Health of Litopenaeus vannamei. Animals (Basel) 2023; 13:2109. [PMID: 37443907 DOI: 10.3390/ani13132109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
The study aimed to assess the impact of adding chenodeoxycholic acid (CDCA) to the diet of Litopenaeus vannamei on their growth performance, lipid and cholesterol metabolism, and hepatopancreas health while being fed a low fishmeal diet. Five diets were formulated, one of which contained 25% fishmeal (PC); fishmeal was partially replaced with Clostridium autoethanogenum protein in the remaining four diets and supplemented with 0, 0.03, 0.06, and 0.09% CDCA (NC, BA1, BA2, and BA3, respectively). In this study, four replicates of each diet were assigned and each replicate consisted of 30 shrimp with an average weight of (0.25 ± 0.03 g). The shrimp were fed four times a day for a period of 56 days. The results of this study indicate that the inclusion of CDCA in the diet had a positive impact on the growth performance of the shrimp. The final body weight (FBW), weight gain (WG), and specific growth rate (SGR) of the shrimp in the PC group were similar to those in the BA2 group, and significantly higher than those in the other three groups. The survival rate (SR) was similar among all groups. In comparison to the PC group, the low fishmeal groups exhibited a significant decrease in the crude lipid content of the whole shrimp, as well as the Total cholesterol (T-CHOL), Low-density lipoprotein (LDL-C), and High-density lipoprotein (HDL-C) levels in the hemolymph. Regarding the sterol metabolism, the dietary supplementation of CDCA up-regulated the mRNA expression of intracellular cholesterol transporter 1-like (npc1), 7-dehydrocholesterol reductase (7dhcr), Delta (24) sterol reductase (Δ24), HMG-CoA reductase membrane form (hmgcr), and sterol carrier protein 2 (scp). In the lipid metabolism, the mRNA expression of sterol-regulatory element binding protein (srebp) was significantly down-regulated in the shrimp fed the BA1 diet and the expression of AMP-activated protein kinase (ampk) was significantly up-regulated in the shrimp fed the BA1 and BA3 diets compared to the PC group. The mRNA expression of triacylglycerol lipase (tgl) was significantly up-regulated in the shrimp fed the BA2 diet compared to the NC group. Compared with the shrimp fed the PC diets, the dietary supplementation of CDCA significantly down-regulated the protein expression of SREBP1. The lumen damage in the BA1 group was significantly less severe than those in the NC group. The addition of 0.06% CDCA to low fishmeal diets can improve the growth performance, lipid and cholesterol metabolism, and hepatopancreas health of L. vannamei.
Collapse
Affiliation(s)
- Menglin Shi
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Chaozhong Zheng
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yidan Sun
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xiaoyue Li
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Guilun He
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Junming Cao
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Zhanjiang 524088, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang 524088, China
| | - Beiping Tan
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Zhanjiang 524088, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang 524088, China
| | - Shiwei Xie
- Laboratory of Aquatic Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Zhanjiang 524088, China
- Key Laboratory of Aquatic, Livestock and Poultry Feed Science and Technology in South China, Ministry of Agriculture, Zhanjiang 524088, China
- Guangdong Provincial Key Lab of Aquatic Animals Disease Control and Healthy Culture, Zhanjiang 524088, China
| |
Collapse
|
11
|
Sawada MIBAC, de Fátima Mello Santana M, Reis M, de Assis SIS, Pereira LA, Santos DR, Nunes VS, Correa-Giannella MLC, Gebrim LH, Passarelli M. Increased plasma lipids in triple-negative breast cancer and impairment in HDL functionality in advanced stages of tumors. Sci Rep 2023; 13:8998. [PMID: 37268673 DOI: 10.1038/s41598-023-35764-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 05/23/2023] [Indexed: 06/04/2023] Open
Abstract
The association between plasma lipids and breast cancer (BC) has been extensively explored although results are still conflicting especially regarding the relationship with high-density lipoprotein cholesterol (HDLc) levels. HDL mediates cholesterol and oxysterol removal from cells limiting sterols necessary for tumor growth, inflammation, and metastasis and this may not be reflected by measuring HDLc. We addressed recently diagnosed, treatment-naïve BC women (n = 163), classified according to molecular types of tumors and clinical stages of the disease, in comparison to control women (CTR; n = 150) regarding plasma lipids and lipoproteins, HDL functionality and composition in lipids, oxysterols, and apo A-I. HDL was isolated by plasma discontinuous density gradient ultracentrifugation. Lipids (total cholesterol, TC; triglycerides, TG; and phospholipids, PL) were determined by enzymatic assays, apo A-I by immunoturbidimetry, and oxysterols (27, 25, and 24-hydroxycholesterol), by gas chromatography coupled with mass spectrometry. HDL-mediated cell cholesterol removal was determined in macrophages previously overloaded with cholesterol and 14C-cholesterol. Lipid profile was similar between CTR and BC groups after adjustment per age. In the BC group, lower concentrations of TC (84%), TG (93%), PL (89%), and 27-hydroxicholesterol (61%) were observed in HDL, although the lipoprotein ability in removing cell cholesterol was similar to HDL from CRT. Triple-negative (TN) BC cases presented higher levels of TC, TG, apoB, and non-HDLc when compared to other molecular types. Impaired HDL functionality was observed in more advanced BC cases (stages III and IV), as cholesterol efflux was around 28% lower as compared to stages I and II. The altered lipid profile in TN cases may contribute to channeling lipids to tumor development in a hystotype with a more aggressive clinical history. Moreover, findings reinforce the dissociation between plasma levels of HDLc and HDL functionality in determining BC outcomes.
Collapse
Affiliation(s)
- Maria Isabela Bloise Alves Caldas Sawada
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
- Centro de Referência da Saúde da Mulher (Hospital Pérola Byington), São Paulo, Brazil
- Hospital da Força Aérea de São Paulo, São Paulo, Brazil
| | - Monique de Fátima Mello Santana
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Mozania Reis
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
- Unidade Básica de Saúde Dra. Ilza Weltman Hutzler, São Paulo, Brazil
| | - Sayonara Ivana Santos de Assis
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Lucas Alves Pereira
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
| | - Danielle Ribeiro Santos
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Valéria Sutti Nunes
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Maria Lucia Cardillo Correa-Giannella
- Laboratório de Carboidratos e Radioimunoensaio Lípides (LIM18), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Henrique Gebrim
- Centro de Referência da Saúde da Mulher (Hospital Pérola Byington), São Paulo, Brazil
| | - Marisa Passarelli
- Programa de Pós-Graduação em Medicina, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil.
- Laboratório de Lípides (LIM10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
12
|
Steck TL, Lange Y. Is reverse cholesterol transport regulated by active cholesterol? J Lipid Res 2023; 64:100385. [PMID: 37169287 PMCID: PMC10279919 DOI: 10.1016/j.jlr.2023.100385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/13/2023] Open
Abstract
This review considers the hypothesis that a small portion of plasma membrane cholesterol regulates reverse cholesterol transport in coordination with overall cellular homeostasis. It appears that almost all of the plasma membrane cholesterol is held in stoichiometric complexes with bilayer phospholipids. The minor fraction of cholesterol that exceeds the complexation capacity of the phospholipids is called active cholesterol. It has an elevated chemical activity and circulates among the organelles. It also moves down its chemical activity gradient to plasma HDL, facilitated by the activity of ABCA1, ABCG1, and SR-BI. ABCA1 initiates this process by perturbing the organization of the plasma membrane bilayer, thereby priming its phospholipids for translocation to apoA-I to form nascent HDL. The active excess sterol and that activated by ABCA1 itself follow the phospholipids to the nascent HDL. ABCG1 similarly rearranges the bilayer and sends additional active cholesterol to nascent HDL, while SR-BI simply facilitates the equilibration of the active sterol between plasma membranes and plasma proteins. Active cholesterol also flows downhill to cytoplasmic membranes where it serves both as a feedback signal to homeostatic ER proteins and as the substrate for the synthesis of mitochondrial 27-hydroxycholesterol (27HC). 27HC binds the LXR and promotes the expression of the aforementioned transport proteins. 27HC-LXR also activates ABCA1 by competitively displacing its inhibitor, unliganded LXR. § Considerable indirect evidence suggests that active cholesterol serves as both a substrate and a feedback signal for reverse cholesterol transport. Direct tests of this novel hypothesis are proposed.
Collapse
Affiliation(s)
- Theodore L Steck
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Yvonne Lange
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
13
|
Zhang Y, Zhou Y, Fang W, Zhu H, Ye C, Zhang D, Lee HJ. Spatial sterol metabolism unveiled by stimulated Raman imaging. Front Chem 2023; 11:1166313. [PMID: 37065823 PMCID: PMC10090450 DOI: 10.3389/fchem.2023.1166313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Graphical AbstractHigh-resolution stimulated Raman scattering (SRS) imaging of a genetically engineered model (GEM) enables metabolite imaging in a yeast model and uncovers an unexpected regulatory mechanism of sterol metabolism, providing new insights underpinning the distributional and functional importance of sterol in cells. SRS-GEM demonstrates a promising platform to explore unknown metabolic mechanisms beyond the reach of conventional approaches.
Collapse
Affiliation(s)
- Yongqing Zhang
- Zhejiang Province Key Laboratory of Quantum Technology and Device, Department of Physics, Interdisciplinary Centre for Quantum Information, Zhejiang University, Hangzhou, China
| | - Yihui Zhou
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Wen Fang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Hanlin Zhu
- Zhejiang Province Key Laboratory of Quantum Technology and Device, Department of Physics, Interdisciplinary Centre for Quantum Information, Zhejiang University, Hangzhou, China
| | - Cunqi Ye
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- *Correspondence: Cunqi Ye, ; Delong Zhang, ; Hyeon Jeong Lee,
| | - Delong Zhang
- Zhejiang Province Key Laboratory of Quantum Technology and Device, Department of Physics, Interdisciplinary Centre for Quantum Information, Zhejiang University, Hangzhou, China
- *Correspondence: Cunqi Ye, ; Delong Zhang, ; Hyeon Jeong Lee,
| | - Hyeon Jeong Lee
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
- *Correspondence: Cunqi Ye, ; Delong Zhang, ; Hyeon Jeong Lee,
| |
Collapse
|
14
|
Zhang M, Liu H, Wang X. Cholesterol oxidase-immobilized MXene/sodium alginate/silica@ n-docosane hierarchical microcapsules for ultrasensitive electrochemical biosensing detection of cholesterol. J Mater Chem B 2023; 11:1506-1522. [PMID: 36655921 DOI: 10.1039/d2tb02367a] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Electrochemical biosensors usually suffer from the deterioration of detection sensitivity and determination accuracy in a high-temperature environment due to protein denaturation and inactivation of their biological recognition elements such as enzymes. Focusing on an effective solution to this crucial issue, we have developed cholesterol oxidase-immobilized MXene/sodium alginate/silica@n-docosane hierarchical microcapsules as a thermoregulatory electrode material for electrochemical biosensors to meet the requirement of ultrasensitive detection of cholesterol at high temperature. The microcapsules were first fabricated by microencapsulating n-docosane as a phase change material (PCM) in a silica shell, followed by depositing a biocompatible sodium alginate layer, wrapping with electroactive MXene nanosheets and then immobilizing cholesterol oxidase as a biological recognition element for electrochemical biosensing. The fabricated composites not only exhibited a layer-by-layer hierarchical microstructure with the desired chemical and biological components, but also obtained a high latent-heat capacity of over 133 J g-1 for thermal management through reversible phase transitions of its PCM core. A bare glassy carbon electrode was modified with the developed composites to serve for the cholesterol biosensor. This enables the modified electrode to obtain an in situ thermoregulatory ability to regulate the microenvironmental temperature surrounding the electrode, effectively preventing the protein denaturation of cholesterol oxidase and minimizing heat impact on biosensing performance. Compared to conventional cholesterol biosensors without a PCM, the developed biosensor achieved a higher sensitivity of 4.63 μA μM-1 cm-2 and a lower limit of detection of 0.081 μM at high temperature, providing highly accurate and reliable detection of cholesterol for real biological samples over a wide temperature range.
Collapse
Affiliation(s)
- Meng Zhang
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Huan Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Xiaodong Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| |
Collapse
|
15
|
Abdul Rashid K, Ibrahim K, Wong JHD, Mohd Ramli N. Lipid Alterations in Glioma: A Systematic Review. Metabolites 2022; 12:metabo12121280. [PMID: 36557318 PMCID: PMC9783089 DOI: 10.3390/metabo12121280] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/08/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Gliomas are highly lethal tumours characterised by heterogeneous molecular features, producing various metabolic phenotypes leading to therapeutic resistance. Lipid metabolism reprogramming is predominant and has contributed to the metabolic plasticity in glioma. This systematic review aims to discover lipids alteration and their biological roles in glioma and the identification of potential lipids biomarker. This systematic review was conducted using the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. Extensive research articles search for the last 10 years, from 2011 to 2021, were conducted using four electronic databases, including PubMed, Web of Science, CINAHL and ScienceDirect. A total of 158 research articles were included in this study. All studies reported significant lipid alteration between glioma and control groups, impacting glioma cell growth, proliferation, drug resistance, patients' survival and metastasis. Different lipids demonstrated different biological roles, either beneficial or detrimental effects on glioma. Notably, prostaglandin (PGE2), triacylglycerol (TG), phosphatidylcholine (PC), and sphingosine-1-phosphate play significant roles in glioma development. Conversely, the most prominent anti-carcinogenic lipids include docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and vitamin D3 have been reported to have detrimental effects on glioma cells. Furthermore, high lipid signals were detected at 0.9 and 1.3 ppm in high-grade glioma relative to low-grade glioma. This evidence shows that lipid metabolisms were significantly dysregulated in glioma. Concurrent with this knowledge, the discovery of specific lipid classes altered in glioma will accelerate the development of potential lipid biomarkers and enhance future glioma therapeutics.
Collapse
Affiliation(s)
- Khairunnisa Abdul Rashid
- Department of Biomedical Imaging, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Kamariah Ibrahim
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Jeannie Hsiu Ding Wong
- Department of Biomedical Imaging, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Norlisah Mohd Ramli
- Department of Biomedical Imaging, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Correspondence: ; Tel.: +60-379673238
| |
Collapse
|
16
|
Saito H, Tachiura W, Nishimura M, Shimizu M, Sato R, Yamauchi Y. Hydroxylation site-specific and production-dependent effects of endogenous oxysterols on cholesterol homeostasis: Implications for SREBP-2 and LXR. J Biol Chem 2022; 299:102733. [PMID: 36423680 PMCID: PMC9792893 DOI: 10.1016/j.jbc.2022.102733] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 10/26/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022] Open
Abstract
The cholesterol metabolites, oxysterols, play central roles in cholesterol feedback control. They modulate the activity of two master transcription factors that control cholesterol homeostatic responses, sterol regulatory element-binding protein-2 (SREBP-2) and liver X receptor (LXR). Although the role of exogenous oxysterols in regulating these transcription factors has been well established, whether endogenously synthesized oxysterols similarly control both SREBP-2 and LXR remains poorly explored. Here, we carefully validate the role of oxysterols enzymatically synthesized within cells in cholesterol homeostatic responses. We first show that SREBP-2 responds more sensitively to exogenous oxysterols than LXR in Chinese hamster ovary cells and rat primary hepatocytes. We then show that 25-hydroxycholesterol (25-HC), 27-hydroxycholesterol, and 24S-hydroxycholesterol endogenously synthesized by CH25H, CYP27A1, and CYP46A1, respectively, suppress SREBP-2 activity at different degrees by stabilizing Insig (insulin-induced gene) proteins, whereas 7α-hydroxycholesterol has little impact on SREBP-2. These results demonstrate the role of site-specific hydroxylation of endogenous oxysterols. In contrast, the expression of CH25H, CYP46A1, CYP27A1, or CYP7A1 fails to induce LXR target gene expression. We also show the 25-HC production-dependent suppression of SREBP-2 using a tetracycline-inducible CH25H expression system. To induce 25-HC production physiologically, murine macrophages are stimulated with a Toll-like receptor 4 ligand, and its effect on SREBP-2 and LXR is examined. The results also suggest that de novo synthesis of 25-HC preferentially regulates SREBP-2 activity. Finally, we quantitatively determine the specificity of the four cholesterol hydroxylases in living cells. Based on our current findings, we conclude that endogenous side-chain oxysterols primarily regulate the activity of SREBP-2, not LXR.
Collapse
Affiliation(s)
- Hodaka Saito
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Wakana Tachiura
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Mizuki Nishimura
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Makoto Shimizu
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryuichiro Sato
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan,Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Yoshio Yamauchi
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan,Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan,For correspondence: Yoshio Yamauchi
| |
Collapse
|
17
|
Xu H, Zheng LX, Chen XS, Pang QY, Yan YN, Liu R, Guo HM, Ren ZY, Yang Y, Gu ZY, Gao C, Gao Y, Luo CL, Zhao Y, Wang Y, Wang T, Tao LY. Brain-specific loss of Abcg1 disturbs cholesterol metabolism and aggravates pyroptosis and neurological deficits after traumatic brain injury. Brain Pathol 2022; 33:e13126. [PMID: 36271611 PMCID: PMC10154369 DOI: 10.1111/bpa.13126] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/04/2022] [Indexed: 11/29/2022] Open
Abstract
Based on accumulating evidence, cholesterol metabolism dysfunction has been suggested to contribute to the pathophysiological process of traumatic brain injury (TBI) and lead to neurological deficits. As a key transporter of cholesterol that efflux from cells, the ATP-binding cassette (ABC) transporter family exerts many beneficial effects on central nervous system (CNS) diseases. However, there is no study regarding the effects and mechanisms of ABCG1 on TBI. As expected, TBI resulted in the different time-course changes of cholesterol metabolism-related molecules in the injured cortex. Considering ABCG1 is expressed in neuron and glia post-TBI, we generated nestin-specific Abcg1 knockout (Abcg1-KO) mice using the Cre/loxP recombination system. These Abcg1-KO mice showed reduced plasma high-density lipoprotein cholesterol levels and increased plasma lower-density lipoprotein cholesterol levels under the base condition. After TBI, these Abcg1-KO mice were susceptible to cholesterol metabolism turbulence. Moreover, Abcg1-KO exacerbated TBI-induced pyroptosis, apoptosis, neuronal cell insult, brain edema, neurological deficits, and brain lesion volume. Importantly, we found that treating with retinoid X receptor (RXR, the upstream molecule of ABCG1) agonist, bexarotene, in Abcg1-KO mice partly rescued TBI-induced neuronal damages mentioned above and improved functional deficits versus vehicle-treated group. These data show that, in addition to regulating brain cholesterol metabolism, Abcg1 improves neurological deficits through inhibiting pyroptosis, apoptosis, neuronal cell insult, and brain edema. Moreover, our findings demonstrate that the cerebroprotection of Abcg1 on TBI partly relies on the activation of the RXRalpha/PPARgamma pathway, which provides a potential therapeutic target for treating TBI.
Collapse
Affiliation(s)
- Heng Xu
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Le-Xin Zheng
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Xue-Shi Chen
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Qiu-Yu Pang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Ya-Nan Yan
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Rong Liu
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Han-Mu Guo
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Zhi-Yang Ren
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Yan Yang
- Department of Pathology and Pathophysiology, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Zhi-Ya Gu
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Cheng Gao
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Yuan Gao
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Cheng-Liang Luo
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Ying Zhao
- Department of Pathology and Pathophysiology, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Ying Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Tao Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Lu-Yang Tao
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| |
Collapse
|
18
|
Perez-Salas U, Porcar L, Garg S, Ayee MAA, Levitan I. Effective Parameters Controlling Sterol Transfer: A Time-Resolved Small-Angle Neutron Scattering Study. J Membr Biol 2022; 255:423-435. [PMID: 35467109 DOI: 10.1007/s00232-022-00231-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 03/19/2022] [Indexed: 11/29/2022]
Abstract
Though cholesterol is the most prevalent and essential sterol in mammalian cellular membranes, its precursors, post-synthesis cholesterol products, as well as its oxidized derivatives play many other important physiological roles. Using a non-invasive in situ technique, time-resolved small angle neutron scattering, we report on the rate of membrane desorption and corresponding activation energy for this process for a series of sterol precursors and post-synthesis cholesterol products that vary from cholesterol by the number and position of double bonds in B ring of cholesterol's steroid core. In addition, we report on sterols that have oxidation modifications in ring A and ring B of the steroid core. We find that sterols that differ in position or the number of double bonds in ring B have similar time and energy characteristics, while oxysterols have faster transfer rates and lower activation energies than cholesterol in a manner generally consistent with known sterol characteristics, like Log P, the n-octanol/water partitioning coefficient. We find, however, that membrane/water partitioning which is dependent on lipid-sterol interactions is a better predictor, shown by the correlation of the sterols' tilt modulus with both the desorption rates and activation energy.
Collapse
Affiliation(s)
- Ursula Perez-Salas
- Physics Department, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| | - Lionel Porcar
- Institut Laue Langevin, 71 Avenue des Martyrs, 38042, Grenoble Cedex 9, France
| | - Sumit Garg
- Physics Department, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Manuela A A Ayee
- Department of Engineering, Dordt University, Sioux Center, IA, USA
| | - Irena Levitan
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60607, USA
| |
Collapse
|
19
|
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: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/09/2022] [Accepted: 07/17/2022] [Indexed: 02/07/2023]
|
20
|
Aguilar-Cazares D, Chavez-Dominguez R, Marroquin-Muciño M, Perez-Medina M, Benito-Lopez JJ, Camarena A, Rumbo-Nava U, Lopez-Gonzalez JS. The systemic-level repercussions of cancer-associated inflammation mediators produced in the tumor microenvironment. Front Endocrinol (Lausanne) 2022; 13:929572. [PMID: 36072935 PMCID: PMC9441602 DOI: 10.3389/fendo.2022.929572] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/01/2022] [Indexed: 12/15/2022] Open
Abstract
The tumor microenvironment is a dynamic, complex, and redundant network of interactions between tumor, immune, and stromal cells. In this intricate environment, cells communicate through membrane-membrane, ligand-receptor, exosome, soluble factors, and transporter interactions that govern cell fate. These interactions activate the diverse and superfluous signaling pathways involved in tumor promotion and progression and induce subtle changes in the functional activity of infiltrating immune cells. The immune response participates as a selective pressure in tumor development. In the early stages of tumor development, the immune response exerts anti-tumor activity, whereas during the advanced stages, the tumor establishes mechanisms to evade the immune response, eliciting a chronic inflammation process that shows a pro-tumor effect. The deregulated inflammatory state, in addition to acting locally, also triggers systemic inflammation that has repercussions in various organs and tissues that are distant from the tumor site, causing the emergence of various symptoms designated as paraneoplastic syndromes, which compromise the response to treatment, quality of life, and survival of cancer patients. Considering the tumor-host relationship as an integral and dynamic biological system, the chronic inflammation generated by the tumor is a communication mechanism among tissues and organs that is primarily orchestrated through different signals, such as cytokines, chemokines, growth factors, and exosomes, to provide the tumor with energetic components that allow it to continue proliferating. In this review, we aim to provide a succinct overview of the involvement of cancer-related inflammation at the local and systemic level throughout tumor development and the emergence of some paraneoplastic syndromes and their main clinical manifestations. In addition, the involvement of these signals throughout tumor development will be discussed based on the physiological/biological activities of innate and adaptive immune cells. These cellular interactions require a metabolic reprogramming program for the full activation of the various cells; thus, these requirements and the by-products released into the microenvironment will be considered. In addition, the systemic impact of cancer-related proinflammatory cytokines on the liver-as a critical organ that produces the leading inflammatory markers described to date-will be summarized. Finally, the contribution of cancer-related inflammation to the development of two paraneoplastic syndromes, myelopoiesis and cachexia, will be discussed.
Collapse
Affiliation(s)
- Dolores Aguilar-Cazares
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Rodolfo Chavez-Dominguez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Mario Marroquin-Muciño
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Laboratorio de Quimioterapia Experimental, Departamento de Bioquimica, Escuela Nacional de Ciencias Biologicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Mario Perez-Medina
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Laboratorio de Quimioterapia Experimental, Departamento de Bioquimica, Escuela Nacional de Ciencias Biologicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Jesus J. Benito-Lopez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Angel Camarena
- Laboratorio de Human Leukocyte Antigen (HLA), Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Uriel Rumbo-Nava
- Clinica de Neumo-Oncologia, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Jose S. Lopez-Gonzalez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| |
Collapse
|
21
|
Messedi M, Guidara W, Grayaa S, Khrouf W, Snoussi M, Bahloul Z, Bonnefont-Rousselot D, Lamari F, Ayadi F. Selected plasma oxysterols as a potential multi-marker biosignature panel for Behçet's Disease. J Steroid Biochem Mol Biol 2022; 221:106122. [PMID: 35588947 DOI: 10.1016/j.jsbmb.2022.106122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
Clinical, genetic, and medical evidence has shown the inflammatory vasculitis aspect of Behçet's Disease (BD). Whereas oxysterols are vital factors in inflammation and oxidative stress, it is still unknown whether they are involved in the pathophysiology of BD. The current study aims to explore the profile of oxysterols in plasma of BD patients. Thirty patients diagnosed with BD and forty healthy controls matched for age and gender were included. Results showed that the cholestane-3β,5α,6β-triol, 27-hydroxycholesterol (27-OHC) and cholestanol levels were higher in BD than controls. In addition, plasma levels of 7-ketocholesterol (7-KC) and 25-hydroxycholesterol (25-OHC) were lower in BD patient. However, levels of 24S-hydroxycholesterol (24-OHC) did not significantly differ. For BD patients, the plasma 7-KC level was negatively correlated with the BD activity index (BDAI) while 27-OHC was positively correlated with high-sensitivity C-reactive protein (hs-CRP) in patients with active course of the disease. According to ROC analysis, a remarkable increase in the area under the curve (AUC) with a higher sensitivity (Se) and specificity (Sp) for 7-KC, 25-OHC and 27-OHC combined markers was observed. The present study indicated that the identification of the predictive value of these three-selected biomarkers related to oxidative stress and inflammation in patients should lead to a better identification of the etiological mechanism of BD.
Collapse
Affiliation(s)
- Meriam Messedi
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Tunisia.
| | - Wassim Guidara
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Tunisia
| | - Sahar Grayaa
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Tunisia
| | - Walid Khrouf
- Service de Biochimie Métabolique, AP-HP.Sorbonne Université, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, DMU BioGeM, Paris F-75013, France
| | - Mouna Snoussi
- Internal medicine department, Hedi Chaker Hosptital, Sfax, Tunisia
| | - Zouhir Bahloul
- Internal medicine department, Hedi Chaker Hosptital, Sfax, Tunisia
| | - Dominique Bonnefont-Rousselot
- Service de Biochimie Métabolique, AP-HP.Sorbonne Université, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, DMU BioGeM, Paris F-75013, France; Université de Paris, CNRS, Inserm, UTCBS, Paris F-75006, France
| | - Foudil Lamari
- Service de Biochimie Métabolique, AP-HP.Sorbonne Université, Hôpitaux Universitaires Pitié-Salpêtrière-Charles Foix, DMU BioGeM, Paris F-75013, France
| | - Fatma Ayadi
- Research Laboratory "Molecular Basis of Human Diseases", LR19ES13, Sfax Medicine School, University of Sfax, Tunisia
| |
Collapse
|
22
|
Suganuma T, Workman JL. MPTAC links alkylation damage signaling to sterol biosynthesis. Redox Biol 2022; 51:102270. [PMID: 35189552 PMCID: PMC8866156 DOI: 10.1016/j.redox.2022.102270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/01/2022] [Accepted: 02/14/2022] [Indexed: 12/19/2022] Open
Abstract
Overproduction of reactive oxygen species (ROS) drives inflammation and mutagenesis. However, the role of the DNA damage response in immune responses remains largely unknown. Here we found that stabilization of the mismatch repair (MMR) protein MSH6 in response to alkylation damage requires interactions with the molybdopterin synthase associating complex (MPTAC) and Ada2a-containing histone acetyltransferase complex (ATAC). Furthermore, MSH6 promotes sterol biosynthesis via the mevalonate pathway in a MPTAC- and ATAC-dependent manner. MPTAC reduces the source of alkylating agents (ROS). Therefore, the association between MMR proteins, MPTAC, and ATAC promotes anti-inflammation response and reduces alkylating agents. The inflammatory responses measured by xanthine oxidase activity are elevated in Lymphoblastoid Cell Lines (LCLs) from some Fragile X-associated disorders (FXD) patients, suggesting that alkylating agents are increased in these FXD patients. However, MPTAC is disrupted in LCLs from some FXD patients. In LCLs from other FXD patients, interaction between MSH6 and ATAC was lost, destabilizing MSH6. Thus, impairment of MPTAC and ATAC may cause alkylation damage resistance in some FXD patients.
Collapse
Affiliation(s)
- Tamaki Suganuma
- Stowers Institute for Medical Research, 1000 E. 50th Street, Kansas City, MO, 64110, USA.
| | - Jerry L Workman
- Stowers Institute for Medical Research, 1000 E. 50th Street, Kansas City, MO, 64110, USA.
| |
Collapse
|
23
|
Lázaro-Mixteco PE, González-Coronel JM, Hernández-Padilla L, Martínez-Alcantar L, Martínez-Carranza E, López-Bucio JS, Guevara-García ÁA, Campos-García J. Transcriptomics Reveals the Mevalonate and Cholesterol Pathways Blocking as Part of the Bacterial Cyclodipeptides Cytotoxic Effects in HeLa Cells of Human Cervix Adenocarcinoma. Front Oncol 2022; 12:790537. [PMID: 35359411 PMCID: PMC8964019 DOI: 10.3389/fonc.2022.790537] [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: 10/07/2021] [Accepted: 02/08/2022] [Indexed: 11/23/2022] Open
Abstract
The incidence of human cervix adenocarcinoma (CC) caused by papillomavirus genome integration into the host chromosome is the third most common cancer among women. Bacterial cyclodipeptides (CDPs) exert cytotoxic effects in human cervical cancer HeLa cells, primarily by blocking the PI3K/Akt/mTOR pathway, but downstream responses comprising gene expression remain unstudied. Seeking to understand the cytotoxic and anti-proliferative effects of CDPs in HeLa cells, a global RNA-Seq analysis was performed. This strategy permitted the identification of 151 differentially expressed genes (DEGs), which were either up- or down-regulated in response to CDPs exposure. Database analysis, including Gene Ontology (COG), and the Kyoto Encyclopedia of Genes and Genomes (KEGG), revealed differential gene expression on cancer transduction signals, and metabolic pathways, for which, expression profiles were modified by the CDPs exposure. Bioinformatics confirmed the impact of CDPs in the differential expression of genes from signal transduction pathways such as PI3K-Akt, mTOR, FoxO, Wnt, MAPK, P53, TGF-β, Notch, apoptosis, EMT, and CSC. Additionally, the CDPs exposure modified the expression of cancer-related transcription factors involved in the regulation of processes such as epigenetics, DNA splicing, and damage response. Interestingly, transcriptomic analysis revealed the participation of genes of the mevalonate and cholesterol biosynthesis pathways; in agreement with this observation, total cholesterol diminished, confirming the blockage of the cholesterol synthesis by the exposure of HeLa cells to CDPs. Interestingly, the expression of some genes of the mevalonate and cholesterol synthesis such as HMGS1, HMGCR, IDI1, SQLE, MSMO1, SREBF1, and SOAT1 was up-regulated by CDPs exposure. Accordingly, metabolites of the mevalonate pathway were accumulated in cultures treated with CDPs. This finding further suggests that the metabolism of cholesterol is crucial for the occurrence of CC, and the blockade of the sterol synthesis as an anti-proliferative mechanism of the bacterial CDPs, represents a reasonable chemotherapeutic drug target to explore. Our transcriptomic study supports the anti-neoplastic effects of bacterial CDPs in HeLa cells shown previously, providing new insights into the transduction signals, transcription factors and metabolic pathways, such as mevalonate and cholesterol that are impacted by the CDPs and highlights its potential as anti-neoplastic drugs.
Collapse
Affiliation(s)
- Pedro E Lázaro-Mixteco
- Laboratorio de Biotecnología Microbiana, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - José M González-Coronel
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Laura Hernández-Padilla
- Laboratorio de Biotecnología Microbiana, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Lorena Martínez-Alcantar
- Laboratorio de Biotecnología Microbiana, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Enrique Martínez-Carranza
- Laboratorio de Biotecnología Microbiana, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Jesús Salvador López-Bucio
- CONACYT-UMSNH, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Ángel A Guevara-García
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Jesús Campos-García
- Laboratorio de Biotecnología Microbiana, Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| |
Collapse
|
24
|
Dansey MV, Palavecino Ruiz MD, Ogara MF, Pecci A, Burton G, Alvarez LD. Insights into estrogen receptor alpha modulation by cholestenoic acids. J Steroid Biochem Mol Biol 2022; 217:106046. [PMID: 34920079 DOI: 10.1016/j.jsbmb.2021.106046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 12/01/2022]
Abstract
Oxysterols are a family of over 25 cholesterol metabolites naturally produced by enzymatic or radical oxidation. They are involved in many physiological and pathological pathways. Although their activity has been mainly attributed to the modulation of the Liver X Receptors (LXR), it is currently accepted that oxysterols are quite promiscuous compounds, acting at several targets at the same time. The promiscuity of the oxysterols with the Estrogen Receptor α (ERα) is crucial in several pathologies such as ER+ breast cancer, inflammation and atherosclerosis. Regarding this matter, we have previously reported the synthesis, LXR activity and binding mode of a family of cholestenoic acid analogs with a modified side chain. Here we report the transcriptional activity on the ERα triggered by these compounds and details on the molecular determinants involved in their activities in order to establish structure-activity relationships to shed light over the molecular basis of the promiscuity of these compounds on ER/LXR responses. Our results show that 3β-hydroxy-5-cholestenoic acid can interact with the ERα receptor in a way similar to 26-hydroxycholesterol and is an agonist of the receptor. Using molecular dynamics simulations, we were able to predict the ERα activity of a set of cholestenoic acid analogs with changes in the flexibility and/or steric requirements of the side chain, some of which exhibited selective activation of ERα or LXR.
Collapse
Affiliation(s)
- María V Dansey
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, UMYMFOR, Buenos Aires, Argentina
| | | | - María F Ogara
- CONICET-Universidad de Buenos Aires, IFIBYNE, Buenos Aires, Argentina
| | - Adalí Pecci
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, IFIBYNE, Buenos Aires, Argentina
| | - Gerardo Burton
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Orgánica, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, UMYMFOR, Buenos Aires, Argentina.
| | - Lautaro D Alvarez
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, UMYMFOR, Buenos Aires, Argentina.
| |
Collapse
|
25
|
Li Y, Wang T, Shi HH, Wang YM, Xue CH, Huang QR, Zhang TT. Absorption, Pharmacokinetics, Tissue Distribution, and Excretion Profiles of Sea Cucumber-Derived Sulfated Sterols in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:480-487. [PMID: 34936372 DOI: 10.1021/acs.jafc.1c04218] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Sea cucumber-derived sulfated sterols exhibited more significant bioactivities compared to plant sterols due to the distinctive structure of the sulfate group at the C-3 position; however, their absorption, pharmacokinetics, tissue distribution, and excretion profiles are unknown, which limits the analysis of molecular mechanisms related to their unique activities. In this study, the absorption characteristics of sea cucumber sterols were determined by oral gavage administration, and their pharmacokinetics, excretion, and tissue distribution were studied by tail vein injection. The results showed that SS1 and SS2 reached the peak at 3 h (20.14 ± 1.2 μg/mL) and 4 h (13.32 ± 0.9 μg/mL) in serum, respectively, after oral gavage administration, suggesting the faster absorption rate of SS1 than SS2 due to the difference in the side-chain groups. Besides, lipid-containing food media improved the digestion and absorption rates of sea cucumber sterols. Moreover, SS1 exhibited a relatively longer duration of efficacy than SS2, and they were almost completely excreted within 9 h through urine. Additionally, sea cucumber sterols were found to be mainly accumulated in the liver (P < 0.05), followed by the kidney and spleen. These findings might provide a theoretical basis for the research and development of functional foods and nutraceuticals associated with sea cucumber sterols.
Collapse
Affiliation(s)
- Yue Li
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
| | - Teng Wang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
| | - Hao-Hao Shi
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237 Shandong Province, P. R. China
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237 Shandong Province, P. R. China
| | - Qing-Rong Huang
- Department of Food Science, Rutgers State University, 65 Dudley Road, New Brunswick, New Jersey 08901, United States
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao 266003, P. R. China
| |
Collapse
|
26
|
Mashat RM, Zielinska HA, Holly JMP, Perks CM. A Role for ER-Beta in the Effects of Low-Density Lipoprotein Cholesterol and 27-Hydroxycholesterol on Breast Cancer Progression: Involvement of the IGF Signalling Pathway? Cells 2021; 11:94. [PMID: 35011656 PMCID: PMC8749996 DOI: 10.3390/cells11010094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 02/07/2023] Open
Abstract
Cholesterol-in particular, high levels of low-density lipoprotein (LDL) and its metabolite, 27-hydroxycholesterol (27-OHC)-is correlated with increases in the risks of breast cancer and obesity. Although the high expression of LDL/27-OHC has been reported in breast cancer, its effects and mechanism of action remain to be fully elucidated. In this study, we found that the effects of LDL on cell proliferation were mediated by the activation of the cytochrome P450 enzyme, sterol 27 hydroxylase, and cholesterol 27-hydroxylase (CYP27A1) in both ER-α-positive and ER-α-negative breast cancer cells. We found that treatment with 27-OHC only increased cell growth in oestrogen receptor-α (ER-α)-positive breast cancer cells in an ER-α-dependent manner, but, interestingly, the effects of 27-OHC on cell migration and invasion were independent of ER-α. Using ER-α-negative MDA-MB-231 cells, we found that 27-OHC similarly promoted cell invasion and migration, and this was mediated by oestrogen receptor β (ER-β). These results suggest that 27-OHC promotes breast cancer cell proliferation in ER-α-positive breast cancer cells via ER-α, but migration and invasion are mediated via ER-β in ER-α positive and negative cell lines. The addition of LDL/27OHC increased the production of IGF-I and the abundance of IGF-IR in TNBC. We further found that modulating ER-β using an agonist or antagonist increased or decreased, respectively, levels of the IGF-I and EGF receptors in TNBC. The inhibition of the insulin-like growth factor receptor blocked the effects of cholesterol on cell growth and the migration of TNBC. Using TCGA and METABRIC microarray expression data from invasive breast cancer carcinomas, we also observed that higher levels of ER-beta were associated with higher levels of IGF-IR. Thus, this study shows novel evidence that ER-β is central to the effects of LDL/27OHC on invasion, migration, and the IGF and EGF axes. Our data suggest that targeting ER-β in TNBC could be an alternative approach for downregulating IGF/EGF signalling and controlling the impact of LDL in breast cancer patients.
Collapse
Affiliation(s)
| | | | | | - Claire M. Perks
- IGFs & Metabolic Endocrinology Group, Translational Health Sciences, Bristol Medical School, Learning & Research Building, Southmead Hospital, Bristol BS10 5NB, UK; (R.M.M.); (H.A.Z.); (J.M.P.H.)
| |
Collapse
|
27
|
Larrazabal C, Silva LMR, Pervizaj-Oruqaj L, Herold S, Hermosilla C, Taubert A. P-Glycoprotein Inhibitors Differently Affect Toxoplasma gondii, Neospora caninum and Besnoitia besnoiti Proliferation in Bovine Primary Endothelial Cells. Pathogens 2021; 10:pathogens10040395. [PMID: 33806177 PMCID: PMC8065907 DOI: 10.3390/pathogens10040395] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Apicomplexan parasites are obligatory intracellular protozoa. In the case of Toxoplasma gondii, Neospora caninum or Besnoitia besnoiti, to ensure proper tachyzoite production, they need nutrients and cell building blocks. However, apicomplexans are auxotrophic for cholesterol, which is required for membrane biosynthesis. P-glycoprotein (P-gp) is a transmembrane transporter involved in xenobiotic efflux. However, the physiological role of P-gp in cholesterol metabolism is unclear. Here, we analyzed its impact on parasite proliferation in T. gondii-, N. caninum- and B. besnoiti-infected primary endothelial cells by applying different generations of P-gp inhibitors. Host cell treatment with verapamil and valspodar significantly diminished tachyzoite production in all three parasite species, whereas tariquidar treatment affected proliferation only in B. besnoiti. 3D-holotomographic analyses illustrated impaired meront development driven by valspodar treatment being accompanied by swollen parasitophorous vacuoles in the case of T. gondii. Tachyzoite and host cell pre-treatment with valspodar affected infection rates in all parasites. Flow cytometric analyses revealed verapamil treatment to induce neutral lipid accumulation. The absence of a pronounced anti-parasitic impact of tariquidar, which represents here the most selective P-gp inhibitor, suggests that the observed effects of verapamil and valspodar are associated with mechanisms independent of P-gp. Out of the three species tested here, this compound affected only B. besnoiti proliferation and its effect was much milder as compared to verapamil and valspodar.
Collapse
Affiliation(s)
- Camilo Larrazabal
- Biomedical Research Center Seltersberg, Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (L.M.R.S.); (C.H.); (A.T.)
- Correspondence:
| | - Liliana M. R. Silva
- Biomedical Research Center Seltersberg, Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (L.M.R.S.); (C.H.); (A.T.)
| | - Learta Pervizaj-Oruqaj
- The Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; (L.P.-O.); (S.H.)
- Member of the German Center for Lung Research (DZL), Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig University Giessen, 35392 Giessen, Germany
| | - Susanne Herold
- The Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; (L.P.-O.); (S.H.)
- Member of the German Center for Lung Research (DZL), Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig University Giessen, 35392 Giessen, Germany
| | - Carlos Hermosilla
- Biomedical Research Center Seltersberg, Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (L.M.R.S.); (C.H.); (A.T.)
| | - Anja Taubert
- Biomedical Research Center Seltersberg, Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (L.M.R.S.); (C.H.); (A.T.)
| |
Collapse
|
28
|
Vona R, Iessi E, Matarrese P. Role of Cholesterol and Lipid Rafts in Cancer Signaling: A Promising Therapeutic Opportunity? Front Cell Dev Biol 2021; 9:622908. [PMID: 33816471 PMCID: PMC8017202 DOI: 10.3389/fcell.2021.622908] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Cholesterol is a lipid molecule that plays an essential role in a number of biological processes, both physiological and pathological. It is an essential structural constituent of cell membranes, and it is fundamental for biosynthesis, integrity, and functions of biological membranes, including membrane trafficking and signaling. Moreover, cholesterol is the major lipid component of lipid rafts, a sort of lipid-based structures that regulate the assembly and functioning of numerous cell signaling pathways, including those related to cancer, such as tumor cell growth, adhesion, migration, invasion, and apoptosis. Considering the importance of cholesterol metabolism, its homeostasis is strictly regulated at every stage: import, synthesis, export, metabolism, and storage. The alterations of this homeostatic balance are known to be associated with cardiovascular diseases and atherosclerosis, but mounting evidence also connects these behaviors to increased cancer risks. Although there is conflicting evidence on the role of cholesterol in cancer development, most of the studies consistently suggest that a dysregulation of cholesterol homeostasis could lead to cancer development. This review aims to discuss the current understanding of cholesterol homeostasis in normal and cancerous cells, summarizing key findings from recent preclinical and clinical studies that have investigated the role of major players in cholesterol regulation and the organization of lipid rafts, which could represent promising therapeutic targets.
Collapse
Affiliation(s)
- Rosa Vona
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità [Italian National Institute of Health], Rome, Italy
| | - Elisabetta Iessi
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità [Italian National Institute of Health], Rome, Italy
| | - Paola Matarrese
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità [Italian National Institute of Health], Rome, Italy
| |
Collapse
|
29
|
Li W, Yang C, Mei X, Huang R, Zhang S, Tang Y, Dong Q, Zhou C. Effect of the polyphenol-rich extract from Allium cepa on hyperlipidemic sprague-dawley rats. J Food Biochem 2020; 45:e13565. [PMID: 33219537 DOI: 10.1111/jfbc.13565] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 10/31/2020] [Accepted: 11/01/2020] [Indexed: 12/13/2022]
Abstract
Allium cepa is used for the prevention and treatment of hyperlipidemia-related diseases such as atherosclerosis in the folk. This study was mainly aimed at investigating the effects of A. cepa extract (ACE) enriched in polyphenols on hyperlipidemia Sprague-Dawley (SD) experiment rat models. The levels of total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), malondialdehyde (MDA), and superoxide dismutase (SOD) activity in serum and liver were measured using ELISA kits. In addition, hematoxylin-eosin (HE) technique was used to observe the liver and the aortic arch pathology. Moreover, western blotting (WB) method was applied to analyze LDL receptor (LDLR) and 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase (HMGCR) in liver. As a result, quercetin (2.42 mg/g DW) and isoquercitrin (4.60 mg/g DW) were the main constituents of ACE using HPLC analysis. Furthermore, ACE reduced the levels of TC, TG, LDL-C, and MDA, and increased HDL levels and elevated SOD activity both in serum and liver in hyperlipidemic SD rats (p < .05). HE results showed that liver fat drops of the rats in ACE group were obviously decreased, and the lipid and foam cells of the aortic arch of the rats in ACE group were markedly ameliorated. WB results showed that ACE promoted the degradation of HMGCR and increased LDLR expression in liver (p < .05). In conclusion, ACE alleviated hyperlipidemia with downregulation of HMGCR and upregulation of LDLR. PRACTICAL APPLICATIONS: Atherosclerosis, a major cardiovascular disease, is the leading cause of mortality and morbidity in the developed countries. Moreover, accumulating data indicate that, during atherosclerosis development, hyperlipidemia is an important risk factor. To date, hyperlipidemia is mainly treated with hyperlipidemic agents including statins, in spite of the side effects and poor tolerance in some patients. In addition, Allium cepa is a medicinal and edible plant. Furthermore, A. cepa is used for the prevention and treatment of hyperlipidemia-related diseases such as atherosclerosis in the folk. But the underlying mechanism is still unclear. In fact, this research showed that A. cepa extract (ACE) alleviated hyperlipidemia with downregulation of HMGCR and upregulation of LDLR, suggesting that ACE might be a potential option for hyperlipidemia as non-statin lipid-lowering agent.
Collapse
Affiliation(s)
- Wenyi Li
- School of Pharmacy, Institute of Material Medica, North Sichuan Medical College, Nanchong, China
| | - Chunyan Yang
- School of Pharmacy, Institute of Material Medica, North Sichuan Medical College, Nanchong, China
| | - Xue Mei
- School of Pharmacy, Institute of Material Medica, North Sichuan Medical College, Nanchong, China
| | - Rong Huang
- School of Pharmacy, Institute of Material Medica, North Sichuan Medical College, Nanchong, China
| | - Shipeng Zhang
- School of Pharmacy, Institute of Material Medica, North Sichuan Medical College, Nanchong, China
| | - Yuanyuan Tang
- School of Pharmacy, Institute of Material Medica, North Sichuan Medical College, Nanchong, China
| | - Qiuhong Dong
- School of Pharmacy, Institute of Material Medica, North Sichuan Medical College, Nanchong, China
| | - Chunyang Zhou
- School of Pharmacy, Institute of Material Medica, North Sichuan Medical College, Nanchong, China
| |
Collapse
|
30
|
Kwon YJ, Jang SN, Liu KH, Jung DH. Effect of Korean Red Ginseng on Cholesterol Metabolites in Postmenopausal Women with Hypercholesterolemia: A Pilot Randomized Controlled Trial. Nutrients 2020; 12:nu12113423. [PMID: 33171597 PMCID: PMC7695162 DOI: 10.3390/nu12113423] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/27/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
Korean red ginseng (KRG) is known to exert beneficial effects on cardiovascular health. Meanwhile, reduced estrogen at menopause has been shown to have various adverse impacts on cardiovascular risk factors, including blood lipids. The aim of this pilot study was to investigate the effect of KRG on cholesterol metabolites, which are surrogate markers of cholesterol absorption and biosynthesis, in postmenopausal women with hypercholesterolemia. The present study is an exploratory study which used data from a 4-week, double-blinded, placebo-controlled clinical pilot study in 68 postmenopausal women with hypercholesterolemia. Patients received KRG (2 g) or placebo (2 g) once daily. The primary endpoints were changes in the levels of nine sterols. Serum sterols were analyzed using liquid chromatography-mass spectrometry (LC-MS)/MS analysis. Among the sterols, reduction in cholesterol level were significantly larger in the KRG group than in the placebo group (the changes: −148.3 ± 261.1 nmol/mL in the ginseng group vs. −23.0 ± 220.5 nmol/mL in the placebo group, p = 0.039). Additionally, changes in 7-hydroxycholesterol (7-OHC) were significantly larger in the KRG group than in the placebo group (the changes: −0.05 ± 0.09 nmol/mL in the ginseng group vs. −0.002 ± 0.1 nmol/mL in the placebo group, p = 0.047). Oxysterols, cholesterol derivates, have been known to play a role in chronic inflammation diseases such as cardiovascular diseases. KRG improves sterol metabolism by decreasing cholesterol and 7-OHC levels in postmenopausal women with hypercholesterolemia.
Collapse
Affiliation(s)
- Yu-Jin Kwon
- Department of Family Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, Korea;
| | - Su-Nyeong Jang
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea;
| | - Kwang-Hyeon Liu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea;
| | - Dong-Hyuk Jung
- Department of Family Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, Korea;
- Correspondence: ; Tel.: +82-10-4204-8998
| |
Collapse
|
31
|
Direct and indirect cholesterol effects on membrane proteins with special focus on potassium channels. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158706. [DOI: 10.1016/j.bbalip.2020.158706] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/19/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022]
|
32
|
Ranjbar Jafarabadi A, Mashjoor S, Mohamadjafari Dehkordi S, Riyahi Bakhtiari A, Cappello T. Steroid Fingerprint Analysis of Endangered Caspian Seal ( Pusa caspica) through the Gorgan Bay (Caspian Sea). ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:7339-7353. [PMID: 32459473 DOI: 10.1021/acs.est.0c01479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The profile of steroid congeners was evaluated in Caspian seals Pusa caspica by age, sex, and tissue-specific bioaccumulation, and compared with that of abiotic matrices (seawater, surface sediment, and suspended particulate materials, SPMs) from Miankaleh Wildlife/Gorgan Bay, (Caspian Sea, Iran). To identify the level of human fecal contamination, ∑25 sterol congeners were measured in all abiotic/biotic samples, revealing coprostanol, a proxy for human feces, as the most abundant sterol (seawater: 45.1-20.3 ng L-1; surface sediment: 90.2-70.3 ng g-1 dw; SPMs: 187.7-157.6 ng g-1 dw). The quantification of ∑25 sterols in seals followed the order of brain > liver > kidney > heart > blood > spleen > muscle > intestine > blubber > fur, and in both sexes coprostanol level (8.95-21.01% of ∑25s) was higher in blubber and fur, followed by cholesterol in brain, liver, kidney, heart, and blood, cholestanone in intestine and muscle, and β-sitosterol in spleen. Though no age/sex differentiation was observed, the mean concentration of ∑25s was higher in male than females and pup. Different diagnostic ratios revealed sterols originating from human and nonhuman sewage sources. Findings pinpoint the urgent necessity to investigate the ecotoxicity of fecal sterols in mammals, and consequent implications for human health.
Collapse
Affiliation(s)
- Ali Ranjbar Jafarabadi
- Department of Environmental Sciences, Faculty of Natural Resources and Marine Sciences, Tarbiat Modares University, Noor, Mazandaran 14115-111, Iran
| | - Sakineh Mashjoor
- Department of Marine biology, Faculty of Marine Science and Technology, University of Hormozgan, Bandar Abbas, Iran
- Marine Pharmaceutical Science Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shirin Mohamadjafari Dehkordi
- Department of Environmental Sciences, Faculty of Natural Resources and Marine Sciences, Tarbiat Modares University, Noor, Mazandaran 14115-111, Iran
| | - Alireza Riyahi Bakhtiari
- Department of Environmental Sciences, Faculty of Natural Resources and Marine Sciences, Tarbiat Modares University, Noor, Mazandaran 14115-111, Iran
| | - Tiziana Cappello
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina 98122, Italy
| |
Collapse
|
33
|
Nam SM, Peterson TA, Butte AJ, Seo KY, Han HW. Explanatory Model of Dry Eye Disease Using Health and Nutrition Examinations: Machine Learning and Network-Based Factor Analysis From a National Survey. JMIR Med Inform 2020; 8:e16153. [PMID: 32130150 PMCID: PMC7059080 DOI: 10.2196/16153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/23/2019] [Accepted: 12/16/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Dry eye disease (DED) is a complex disease of the ocular surface, and its associated factors are important for understanding and effectively treating DED. OBJECTIVE This study aimed to provide an integrative and personalized model of DED by making an explanatory model of DED using as many factors as possible from the Korea National Health and Nutrition Examination Survey (KNHANES) data. METHODS Using KNHANES data for 2012 (4391 sample cases), a point-based scoring system was created for ranking factors associated with DED and assessing patient-specific DED risk. First, decision trees and lasso were used to classify continuous factors and to select important factors, respectively. Next, a survey-weighted multiple logistic regression was trained using these factors, and points were assigned using the regression coefficients. Finally, network graphs of partial correlations between factors were utilized to study the interrelatedness of DED-associated factors. RESULTS The point-based model achieved an area under the curve of 0.70 (95% CI 0.61-0.78), and 13 of 78 factors considered were chosen. Important factors included sex (+9 points for women), corneal refractive surgery (+9 points), current depression (+7 points), cataract surgery (+7 points), stress (+6 points), age (54-66 years; +4 points), rhinitis (+4 points), lipid-lowering medication (+4 points), and intake of omega-3 (0.43%-0.65% kcal/day; -4 points). Among these, the age group 54 to 66 years had high centrality in the network, whereas omega-3 had low centrality. CONCLUSIONS Integrative understanding of DED was possible using the machine learning-based model and network-based factor analysis. This method for finding important risk factors and identifying patient-specific risk could be applied to other multifactorial diseases.
Collapse
Affiliation(s)
- Sang Min Nam
- Department of Ophthalmology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Thomas A Peterson
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, United States
| | - Atul J Butte
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, United States
| | - Kyoung Yul Seo
- Department of Ophthalmology, Institute of Vision Research, Eye and Ear Hospital, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Wook Han
- Department of Biomedical Informatics, CHA University School of Medicine, CHA University, Seongnam, Republic of Korea
| |
Collapse
|
34
|
Parker ME, Barrera A, Wheaton JD, Zuberbuehler MK, Allan DS, Carlyle JR, Reddy TE, Ciofani M. c-Maf regulates the plasticity of group 3 innate lymphoid cells by restraining the type 1 program. J Exp Med 2020; 217:e20191030. [PMID: 31570496 PMCID: PMC7037249 DOI: 10.1084/jem.20191030] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/31/2019] [Accepted: 09/18/2019] [Indexed: 12/30/2022] Open
Abstract
CCR6- group 3 innate lymphoid cells (ILC3s) are mediators of intestinal immunity and barrier function that possess the capacity to acquire type 1 effector features and fully convert into ILC1s. The molecular mechanisms governing such plasticity are undefined. Here, we identified c-Maf as an essential regulator of ILC3 homeostasis and plasticity that limits physiological ILC1 conversion. Phenotypic analysis of effector status in Maf-deficient CCR6- ILC3s, coupled with evaluation of global changes in transcriptome, chromatin accessibility, and transcription factor motif enrichment, revealed that c-Maf enforces ILC3 identity. c-Maf promoted ILC3 accessibility and supported RORγt activity and expression of type 3 effector genes. Conversely, c-Maf antagonized type 1 programming, largely through restraint of T-bet expression and function. Mapping of the dynamic changes in chromatin landscape accompanying CCR6- ILC3 development and ILC1 conversion solidified c-Maf as a gatekeeper of type 1 regulatory transformation and a controller of ILC3 fate.
Collapse
Affiliation(s)
- Morgan E. Parker
- Department of Immunology, Duke University Medical Center, Durham, NC
| | - Alejandro Barrera
- Center for Genomic and Computational Biology, Duke University Medical School, Durham, NC
- Department of Biostatistics and Bioinformatics, Duke University Medical School, Durham, NC
| | - Joshua D. Wheaton
- Department of Immunology, Duke University Medical Center, Durham, NC
| | | | - David S.J. Allan
- Department of Immunology, University of Toronto, Toronto, Canada
| | - James R. Carlyle
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Timothy E. Reddy
- Center for Genomic and Computational Biology, Duke University Medical School, Durham, NC
- Department of Biostatistics and Bioinformatics, Duke University Medical School, Durham, NC
| | - Maria Ciofani
- Department of Immunology, Duke University Medical Center, Durham, NC
| |
Collapse
|
35
|
Pasello M, Giudice AM, Scotlandi K. The ABC subfamily A transporters: Multifaceted players with incipient potentialities in cancer. Semin Cancer Biol 2019; 60:57-71. [PMID: 31605751 DOI: 10.1016/j.semcancer.2019.10.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/30/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022]
Abstract
Overexpression of ATP-binding cassette (ABC) transporters is a cause of drug resistance in a plethora of tumors. More recent evidence indicates additional contribution of these transporters to other processes, such as tumor cell dissemination and metastasis, thereby extending their possible roles in tumor progression. While the role of some ABC transporters, such as ABCB1, ABCC1 and ABCG2, in multidrug resistance is well documented, the mechanisms by which ABC transporters affect the proliferation, differentiation, migration and invasion of cancer cells are still poorly defined and are frequently controversial. This review, summarizes recent advances that highlight the role of subfamily A members in cancer. Emerging evidence highlights the potential value of ABCA members as biomarkers of risk and response in different tumors, but information is disperse and very little is known about their possible mechanisms of action. The only clear evidence is that ABCA members are involved in lipid metabolism and homeostasis. In particular, the relationship between ABCA1 and cholesterol is becoming evident in different fields of biology, including cancer. In parallel, emerging findings indicate that cholesterol, the main component of cell membranes, can influence many physiological and pathological processes, including cell migration, cancer progression and metastasis. This review aims to link the dispersed knowledge regarding the relationship of ABCA members with lipid metabolism and cancer in an effort to stimulate and guide readers to areas that the writers consider to have significant impact and relevant potentialities.
Collapse
Affiliation(s)
- Michela Pasello
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, 40136, Italy.
| | - Anna Maria Giudice
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, 40136, Italy; Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, 40126, Italy
| | - Katia Scotlandi
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, 40136, Italy.
| |
Collapse
|