1
|
Kim J, Yu YS, Choi Y, Lee DH, Han S, Kwon J, Noda T, Ikawa M, Kim D, Kim H, Ballabio A, Kim KI, Baek SH. USF2 and TFEB compete in regulating lysosomal and autophagy genes. Nat Commun 2024; 15:8334. [PMID: 39333072 PMCID: PMC11436898 DOI: 10.1038/s41467-024-52600-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 09/15/2024] [Indexed: 09/29/2024] Open
Abstract
Autophagy, a highly conserved self-digestion process crucial for cellular homeostasis, is triggered by various environmental signals, including nutrient scarcity. The regulation of lysosomal and autophagy-related processes is pivotal to maintaining cellular homeostasis and basal metabolism. The consequences of disrupting or diminishing lysosomal and autophagy systems have been investigated; however, information on the implications of hyperactivating lysosomal and autophagy genes on homeostasis is limited. Here, we present a mechanism of transcriptional repression involving upstream stimulatory factor 2 (USF2), which inhibits lysosomal and autophagy genes under nutrient-rich conditions. We find that USF2, together with HDAC1, binds to the CLEAR motif within lysosomal genes, thereby diminishing histone H3K27 acetylation, restricting chromatin accessibility, and downregulating lysosomal gene expression. Under starvation, USF2 competes with transcription factor EB (TFEB), a master transcriptional activator of lysosomal and autophagy genes, to bind to target gene promoters in a phosphorylation-dependent manner. The GSK3β-mediated phosphorylation of the USF2 S155 site governs USF2 DNA-binding activity, which is involved in lysosomal gene repression. These findings have potential applications in the treatment of protein aggregation-associated diseases, including α1-antitrypsin deficiency. Notably, USF2 repression is a promising therapeutic strategy for lysosomal and autophagy-related diseases.
Collapse
Affiliation(s)
- Jaebeom Kim
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Young Suk Yu
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Yehwa Choi
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Do Hui Lee
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Soobin Han
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Junhee Kwon
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Taichi Noda
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
- Priority Organization for Innovation and Excellence, Kumamoto University, Kumamoto, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Dongha Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyunkyung Kim
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Keun Il Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea.
| | - Sung Hee Baek
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, South Korea.
| |
Collapse
|
2
|
Moss CE, Johnston SA, Kimble JV, Clements M, Codd V, Hamby S, Goodall AH, Deshmukh S, Sudbery I, Coca D, Wilson HL, Kiss-Toth E. Aging-related defects in macrophage function are driven by MYC and USF1 transcriptional programs. Cell Rep 2024; 43:114073. [PMID: 38578825 DOI: 10.1016/j.celrep.2024.114073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/15/2024] [Accepted: 03/21/2024] [Indexed: 04/07/2024] Open
Abstract
Macrophages are central innate immune cells whose function declines with age. The molecular mechanisms underlying age-related changes remain poorly understood, particularly in human macrophages. We report a substantial reduction in phagocytosis, migration, and chemotaxis in human monocyte-derived macrophages (MDMs) from older (>50 years old) compared with younger (18-30 years old) donors, alongside downregulation of transcription factors MYC and USF1. In MDMs from young donors, knockdown of MYC or USF1 decreases phagocytosis and chemotaxis and alters the expression of associated genes, alongside adhesion and extracellular matrix remodeling. A concordant dysregulation of MYC and USF1 target genes is also seen in MDMs from older donors. Furthermore, older age and loss of either MYC or USF1 in MDMs leads to an increased cell size, altered morphology, and reduced actin content. Together, these results define MYC and USF1 as key drivers of MDM age-related functional decline and identify downstream targets to improve macrophage function in aging.
Collapse
Affiliation(s)
- Charlotte E Moss
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Healthy Lifespan Institute, University of Sheffield, Sheffield, UK
| | - Simon A Johnston
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Joshua V Kimble
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Healthy Lifespan Institute, University of Sheffield, Sheffield, UK
| | - Martha Clements
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Veryan Codd
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK; National Institute for Healthcare Research, Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Stephen Hamby
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK; National Institute for Healthcare Research, Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Alison H Goodall
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK; National Institute for Healthcare Research, Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Sumeet Deshmukh
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Ian Sudbery
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Daniel Coca
- Healthy Lifespan Institute, University of Sheffield, Sheffield, UK; Department of Autonomic Control and Systems Engineering, University of Sheffield, Sheffield, UK
| | - Heather L Wilson
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Healthy Lifespan Institute, University of Sheffield, Sheffield, UK.
| | - Endre Kiss-Toth
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Healthy Lifespan Institute, University of Sheffield, Sheffield, UK; Biological Research Centre, Szeged, Hungary.
| |
Collapse
|
3
|
Zheng PF, Chen LZ, Pan HW, Liu P, Zheng ZF. Effects of USF1 SNPs and SNP–Environment Interactions on Serum Lipid Profiles and the Risk of Early-Onset Coronary Artery Disease in the Chinese Population. Front Cardiovasc Med 2022; 9:882728. [PMID: 35783856 PMCID: PMC9240353 DOI: 10.3389/fcvm.2022.882728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundUpstream transcription factor 1 (USF1) single-nucleotide polymorphisms (SNPs) are significantly associated with serum lipid levels in several different ethnic groups or populations, but their association with lipid levels and the risk of early-onset coronary artery disease (EOCAD) has not been reported in Han populations of southern China.MethodsSix USF1 SNPs (rs3737787, rs2774276, rs2516839, rs2516838, rs1556259, and rs2516837) were genotyped by next-generation sequencing (NGS) techniques in 686 control subjects and 728 patients with EOCAD.ResultsThe genotypic and allelic frequencies of the USF1 rs3737787 SNP were significantly different between the control and EOCAD groups. The subgroup analysis identified that the rs3737787T allele was related to a decreased risk of EOCAD, whereas the rs3737787C–rs2774276G–rs2516839A and rs3737787C–rs2774276G–rs2516839G haplotypes were related to an increased risk of EOCAD in men, and the rs3737787C–rs2774276G–rs2516839A and rs3737787T–rs2774276C–rs2516839A haplotypes were correlated with an increased risk of EOCAD in women (p < 0.05–0.01). Male rs3737787T allele carriers had lower low-density lipoprotein cholesterol (LDL-C), total cholesterol (TC), and triglyceride (TG) concentrations than the rs3737787T allele non-carriers (p < 0.01). The interactions of rs3737787 with alcohol consumption and rs2516839 with smoking affected serum TC and LDL-C levels in men, whereas the interaction of rs3737787 with alcohol consumption affected serum high-density lipoprotein cholesterol (HDL-C) levels and the rs2516839-smoking interaction affected serum TC levels in women (pI < 0.001). The expression levels of the USF1 mRNA, interleukin 1β (IL-1β), tumor necrosis factor-α (TNF-α), and interleukin 6 (IL-6) were significantly lower in controls than in patients with EOCAD, and rs3737787T allele carriers displayed lower IL-1β, TNF-α, IL-6, and USF1 mRNA expression levels than the rs3737787T allele non-carriers. In addition, IL-1β, TNF-α, and IL-6 expression levels were significantly positively correlated with USF1 mRNA levels (p < 0.01).ConclusionSex-specific correlations were identified between the USF1 rs3737787T allele with blood lipid levels and the risk of EOCAD. The USF1 rs3737787T allele affects the risk of EOCAD by modulating serum lipid levels and the expression of inflammatory factors, including IL-1β, TNF-α, and IL-6.
Collapse
Affiliation(s)
- Peng-Fei Zheng
- Cardiology Department, Hunan Provincial People's Hospital, Changsha, China
- Clinical Research Center for Heart Failure in Hunan Province, Changsha, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People's Hospital, Changsha, China
| | - Lu-Zhu Chen
- Department of Cardiology, The Central Hospital of ShaoYang, Shaoyang, China
| | - Hong-Wei Pan
- Cardiology Department, Hunan Provincial People's Hospital, Changsha, China
- Clinical Research Center for Heart Failure in Hunan Province, Changsha, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People's Hospital, Changsha, China
| | - Peng Liu
- Department of Cardiology, The Central Hospital of ShaoYang, Shaoyang, China
- *Correspondence: Peng Liu
| | - Zhao-Fen Zheng
- Cardiology Department, Hunan Provincial People's Hospital, Changsha, China
- Clinical Research Center for Heart Failure in Hunan Province, Changsha, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People's Hospital, Changsha, China
- Zhao-Fen Zheng
| |
Collapse
|
4
|
Abstract
Significance: Thioredoxin-interacting protein (Txnip) is an α-arrestin protein that acts as a cancer suppressor. Txnip is simultaneously a critical regulator of energy metabolism. Other alpha-arrestin proteins also play key roles in cell biology and cancer. Recent Advances: Txnip expression is regulated by multilayered mechanisms, including transcriptional regulation, microRNA, messenger RNA (mRNA) stabilization, and protein degradation. The Txnip-based connection between cancer and metabolism has been widely recognized. Meanwhile, new aspects are proposed for the mechanism of action of Txnip, including the regulation of RNA expression and autophagy. Arrestin domain containing 3 (ARRDC3), another α-arrestin protein, regulates endocytosis and signaling, whereas ARRDC1 and ARRDC4 regulate extracellular vesicle formation. Critical Issues: The mechanism of action of Txnip is yet to be elucidated. The regulation of intracellular protein trafficking by arrestin family proteins has opened an emerging field of biology and medical research, which needs to be examined further. Future Directions: A fundamental understanding of the mechanism of action of Txnip and other arrestin family members needs to be explored in the future to combat diseases such as cancer and diabetes. Antioxid. Redox Signal. 36, 1001-1022.
Collapse
Affiliation(s)
- Hiroshi Masutani
- Department of Clinical Laboratory Sciences, Tenri Health Care University, Tenri, Japan.,Department of Infection and Prevention, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
5
|
Abstract
PURPOSE OF REVIEW : Familial combined hyperlipidemia (FCH), defined by concurrently elevated plasma triglyceride (TG) and low-density lipoprotein (LDL) cholesterol, has long been investigated to characterize its genetic basis. Despite almost half a century of searching, a single gene cause for the phenotype has not yet been identified. RECENT FINDINGS : Recent studies using next-generation genetic analytic methods confirm that FCH has a polygenic basis, with a clear large contribution from the accumulation of small-to-moderate effect common single nucleotide polymorphisms (SNPs) throughout the genome that is associated with raising TG, and probably also those raising LDL cholesterol. On the other hand, rare monogenic variants, such as those causing familial hypercholesterolemia, play a negligible role, if any. Genetic profiling suggests that patients with FCH and hypertriglyceridemia share a strong polygenic basis and show a similar profile of multiple TG-raising common SNPs. SUMMARY : Recent progress in genomics has shown that most if not all of the genetic susceptibility to FCH is polygenic in nature. Future research should include larger cohort studies, with wider ancestral diversity, ancestry-specific polygenic scores, and investigation of epigenetic and lifestyle factors to help further elucidate the causative agents at play in cases where the genetic etiology remains to be defined.
Collapse
Affiliation(s)
| | - Robert A Hegele
- Robarts Research Institute
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
6
|
Li Z, Chen X, Tao J, Shi A, Zhang J, Yu P. Exosomes Regulate NLRP3 Inflammasome in Diseases. Front Cell Dev Biol 2022; 9:802509. [PMID: 35047512 PMCID: PMC8762245 DOI: 10.3389/fcell.2021.802509] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/03/2021] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence has suggested the unique and critical role of exosomes as signal molecules vector in various diseases. Numerous researchers have been trying to identify how these exosomes function in immune progression, as this could promote their use as biomarkers for the disease process and potential promising diagnostic tools. NOD-like receptor (NLR) family, pyrin domain containing 3 (NLRP3), a tripartite protein, contains three functional domains a central nucleotide-binding and oligomerization domain (NACHT), an N-terminal pyrin domain (PYD), and a leucine-rich repeat domain (LRR). Of note, existing studies have identified exosome as a novel mediator of the NLRP3 inflammasome, which is critical in diseases progression. However, the actual mechanisms and clinical treatment related to exosomes and NLRP3 are still not fully understood. Herein, we presented an up-to-date review of exosomes and NLRP3 in diseases, outlining what is known about the role of exosomes in the activation of NLRP3 inflammasome and also highlighting areas of this topic that warrant further study.
Collapse
Affiliation(s)
- Zhangwang Li
- The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Xinyue Chen
- The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Junjie Tao
- The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Ao Shi
- School of Medicine, University of Nicosia, Nicosia, Cyprus.,School of Medicine, St. George University of London, London, United Kingdom
| | - Jing Zhang
- The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China.,Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peng Yu
- The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China.,Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
7
|
Abstract
BACKGROUND Glioblastoma stem cells (GSCs) and their interplay with tumor-associated macrophages (TAMs) are responsible for malignant growth and tumor recurrence of glioblastoma multiforme (GBM), but the underlying mechanisms are largely unknown. METHODS Cell viability, stemness, migration, and invasion were measured in GSCs after the knockdown of upstream stimulating factor 1 (USF1). Luciferase assay and chromatin immunoprecipitation qPCR were performed to determine the regulation of CD90 by USF1. Immunohistochemistry and immunofluorescent staining were used to examine the expression of USF1 and GSC markers, as well as the crosstalk between GSCs and TAMs. In addition, the interaction between GSCs and TAMs was confirmed using in vivo GBM models. RESULTS We show that USF1 promotes malignant glioblastoma phenotypes and GSCs-TAMs physical interaction by inducing CD90 expression. USF1 predicts a poor prognosis for glioma patients and is upregulated in patient-derived GSCs and glioblastoma cell lines. USF1 overexpression increases the proliferation, invasion, and neurosphere formation of GSCs and glioblastoma cell lines, while USF1 knockdown exerts an opposite effect. Further mechanistic studies reveal that USF1 promotes GSC stemness by directly regulating CD90 expression. Importantly, CD90 of GSCs functions as an anchor for physical interaction with macrophages. Additionally, the USF1/CD90 signaling axis supports the GSCs and TAMs adhesion and immunosuppressive feature of TAMs, which in turn enhance the stemness of GSCs. Moreover, the overexpression of CD90 restores the stemness property in USF1 knockdown GSCs and its immunosuppressive microenvironment. CONCLUSIONS Our findings indicate that the USF1/CD90 axis might be a potential therapeutic target for the treatment of glioblastoma.
Collapse
|
8
|
Trinder M, Vikulova D, Pimstone S, Mancini GBJ, Brunham LR. Polygenic architecture and cardiovascular risk of familial combined hyperlipidemia. Atherosclerosis 2021; 340:35-43. [PMID: 34906840 DOI: 10.1016/j.atherosclerosis.2021.11.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/06/2021] [Accepted: 11/30/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND AIMS Familial combined hyperlipidemia (FCHL) is one of the most common inherited lipid phenotypes, characterized by elevated plasma concentrations of apolipoprotein B-100 and triglycerides. The genetic inheritance of FCHL remains poorly understood. The goals of this study were to investigate the polygenetic architecture and cardiovascular risk associated with FCHL. METHODS AND RESULTS We identified individuals with an FCHL phenotype among 349,222 unrelated participants of European ancestry in the UK Biobank using modified versions of 5 different diagnostic criteria. The prevalence of the FCHL phenotype was 11.44% (n = 39,961), 5.01% (n = 17,485), 1.48% (n = 5,153), 1.10% (n = 3,838), and 0.48% (n = 1,688) according to modified versions of the Consensus Conference, Dutch, Mexico, Brunzell, and Goldstein criteria, respectively. We performed discovery, case-control genome-wide association studies for these different FCHL criteria and identified 175 independent loci associated with FCHL at genome-wide significance. We investigated the association of genetic and clinical risk with FCHL and found that polygenic susceptibility to hypercholesterolemia or hypertriglyceridemia and features of metabolic syndrome were associated with greater prevalence of FCHL. Participants with an FCHL phenotype had a similar risk of incident coronary artery disease compared to participants with monogenic familial hypercholesterolemia (adjusted hazard ratio vs controls [95% confidence interval]: 2.72 [2.31-3.21] and 1.90 [1.30-2.78]). CONCLUSIONS These results suggest that, rather than being a single genetic entity, the FCHL phenotype represents a polygenic susceptibility to dyslipidemia in combination with metabolic abnormalities. The cardiovascular risk associated with an FCHL phenotype is similar to that of monogenic familial hypercholesterolemia, despite being ∼5x more common.
Collapse
Affiliation(s)
- Mark Trinder
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada; Experimental Medicine Program, University of British Columbia, Vancouver, British Columbia, Canada
| | - Diana Vikulova
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada; Experimental Medicine Program, University of British Columbia, Vancouver, British Columbia, Canada
| | - Simon Pimstone
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada; Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - G B John Mancini
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada
| | - Liam R Brunham
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada; Experimental Medicine Program, University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
9
|
Genetics of Familial Combined Hyperlipidemia (FCHL) Disorder: An Update. Biochem Genet 2021; 60:453-481. [PMID: 34478023 DOI: 10.1007/s10528-021-10130-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 08/09/2021] [Indexed: 10/20/2022]
Abstract
Familial combined hyperlipidemia (FCHL) is one of the most common familial lipoprotein disorders of the lipoproteins, with a prevalence of 0.5% to 2% in different populations. About 10% of these patients suffer from cardiovascular disease and this number is increased by up to 11.3% in the young survivors of myocardial infarction and by 40% among all the survivors of myocardial infarction. Although initially thought to be that FCHL has an inheritance pattern of monogenic, the disease's etiology is still not fully understood and it appears that FCHL has a complex pattern related to genetic variants, environmental factors, and lifestyles. Two strategies have been used to identify its complex genetic background: candidate gene and the linkage approach, which have yielded an extensive list of genes associated with FCHL with a variable degree of scientific evidence. Until now, more than 30 different genetic variants have been identified related to FCHL. In this study, we aimed to review the individual genes that have been described in FCHL and how these genes and variants can be related to the current concept of metabolic pathways resulting in familial combined hyperlipidemia.
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Hypertriglyceridemia is a common dyslipidemia associated with an increased risk of cardiovascular disease and pancreatitis. Severe hypertriglyceridemia may sometimes be a monogenic condition. However, in the vast majority of patients, hypertriglyceridemia is due to the cumulative effect of multiple genetic risk variants along with lifestyle factors, medications, and disease conditions that elevate triglyceride levels. In this review, we will summarize recent progress in the understanding of the genetic basis of hypertriglyceridemia. RECENT FINDINGS More than 300 genetic loci have been identified for association with triglyceride levels in large genome-wide association studies. Studies combining the loci into polygenic scores have demonstrated that some hypertriglyceridemia phenotypes previously attributed to monogenic inheritance have a polygenic basis. The new genetic discoveries have opened avenues for the development of more effective triglyceride-lowering treatments and raised interest towards genetic screening and tailored treatments against hypertriglyceridemia. The discovery of multiple genetic loci associated with elevated triglyceride levels has led to improved understanding of the genetic basis of hypertriglyceridemia and opened new translational opportunities.
Collapse
Affiliation(s)
- Germán D. Carrasquilla
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Mærsk Building, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Malene Revsbech Christiansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Mærsk Building, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Tuomas O. Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Mærsk Building, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| |
Collapse
|
11
|
Pedro-Botet J, Climent E, Gabarró N, Millán J. Familial combined hyperlipidaemia/polygenic mixed hyperlipidaemia. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2021; 33 Suppl 2:43-49. [PMID: 34006353 DOI: 10.1016/j.arteri.2020.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 12/14/2020] [Indexed: 11/26/2022]
Abstract
Familial combined hyperlipidaemia (FCH) is the most prevalent form of familial hyperlipidaemia with a multigenic origin and a complex pattern of inheritance. In this respect, FCH is an oligogenic primary lipid disorder due to interaction of genetic variants and mutations with environmental factors. Patients with FCH are at increased risk of cardiovascular disease and often have other associated metabolic conditions. Despite its relevance in cardiovascular prevention, FCH is frequently underdiagnosed and very often undertreated. In this review, emphasis is placed on the most recent advances in FCH, in order to increase its awareness and ultimately contribute to improving its clinical control.
Collapse
Affiliation(s)
- Juan Pedro-Botet
- Unidad de Lípidos y Riesgo Vascular, Hospital del Mar, Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, España.
| | - Elisenda Climent
- Unidad de Lípidos y Riesgo Vascular, Hospital del Mar, Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, España
| | - Nuria Gabarró
- Unidad de Lípidos, Servicio de Medicina Interna, H. U. Gregorio Marañón, Universidad Complutense, Madrid, España
| | - Jesús Millán
- Unidad de Lípidos, Servicio de Medicina Interna, H. U. Gregorio Marañón, Universidad Complutense, Madrid, España
| |
Collapse
|
12
|
Inamoto I, Sheoran I, Popa SC, Hussain M, Shin JA. Combining Rational Design and Continuous Evolution on Minimalist Proteins That Target the E-box DNA Site. ACS Chem Biol 2021; 16:35-44. [PMID: 33370105 DOI: 10.1021/acschembio.0c00684] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Protein-based therapeutics are part of the next-generation arsenal of drugs being developed against proto-oncoprotein Myc. We designed protein MEF to mimic the basic region/helix-loop-helix/leucine zipper (bHLHZ) domain of Max and Myc, which bind to the E-box motif (enhancer box, CACGTG). To make MEF, we started with our rationally designed ME47, a hybrid of the Max basic region and E47 HLH, that effectively inhibited tumor growth in a mouse model of breast cancer. We used phage-assisted continuous evolution (PACE), which uncovered mutations at Arg12 that contact the DNA phosphodiester backbone. The Arg12 mutations improved ME47's stability. We replaced Cys29 with Ala to eliminate potential undesired disulfide formation and fused the designed FosW leucine zipper to mutated ME47 to increase the dimerization interface and E-box targeting activity. This "franken-protein" MEF comprises the Max basic region, E47 HLH, and FosW leucine zipper. Compared with ME47, MEF gives 2-fold stronger binding to E-box and 4-fold increased specificity for E-box over nonspecific DNA. The synergistic combination of rational design and PACE allowed us to make MEF and demonstrates the power and utility of our two-pronged approach toward development of promising protein drugs with robust structure and DNA-binding function.
Collapse
Affiliation(s)
- Ichiro Inamoto
- Department of Chemistry, University of Toronto, 3359 Mississauga Road, Mississauga, ON L5L 1C6, Canada
| | - Inder Sheoran
- Department of Chemistry, University of Toronto, 3359 Mississauga Road, Mississauga, ON L5L 1C6, Canada
| | - Serban C. Popa
- Department of Chemistry, University of Toronto, 3359 Mississauga Road, Mississauga, ON L5L 1C6, Canada
| | - Montdher Hussain
- Department of Chemistry, University of Toronto, 3359 Mississauga Road, Mississauga, ON L5L 1C6, Canada
| | - Jumi A. Shin
- Department of Chemistry, University of Toronto, 3359 Mississauga Road, Mississauga, ON L5L 1C6, Canada
| |
Collapse
|
13
|
Polymorphism rs3737787 of Upstream Stimulatory Factor 1 gene is associated with serum lipid phenotype in Nigerian population. Mol Cell Probes 2020; 55:101687. [PMID: 33307180 DOI: 10.1016/j.mcp.2020.101687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/22/2020] [Accepted: 12/01/2020] [Indexed: 11/21/2022]
Abstract
Serum lipid profile which is determined by genotype-phenotype relationship plays a significant role in the development of cardiovascular disease. Upstream stimulatory factor 1 (USF1), has been reported to be associated with serum lipid levels in different population, hence, this study investigated the association of variants in USF1 with serum lipid profile in adults in Lagos state, Nigeria. We genotyped rs3737787 (11235C > T) and rs550376620 (10488G > A) with PCR-RFLP in 384 participants and we used logistic regression to assess the association of these variants with serum lipid levels. The minor allele frequency observed in 10488G > A in both case and control groups was 5% while the minor allele of 11235C > T was observed to be more frequent in the control when compared to the dyslipidemic subjects (24% vs 12%; p = 1.84e-05). Levels of total cholesterol, triglycerides, and LDL-c in dyslipidemic subjects with CC genotype of 11235C > T were significantly higher compared to CT and TT genotypes (p < 0.001; p < 0.0001 and p < 0.0001 respectively). Logistic regression with adjustment for age, gender and BMI, showed that the minor allele carriers of 11235C > T have a reduced risk of dyslipidemia (Odds ratio: 0. 0.043, 95% confidence interval (CI): (0.006-0.331, p = 0.002). Our findings revealed that rs3737787 is associated with lipid phenotype in Nigerian population.
Collapse
|
14
|
Abstract
Hypertriglyceridemia is one of the most common lipid abnormalities encountered in clinical practice. Many monogenic disorders causing severe hypertriglyceridemia have been identified, but in most patients triglyceride elevations result from a combination of multiple genetic variations with small effects and environmental factors. Common secondary causes include obesity, uncontrolled diabetes, alcohol misuse, and various commonly used drugs. Correcting these factors and optimizing lifestyle choices, including dietary modification, is important before starting drug treatment. The goal of drug treatment is to reduce the risk of pancreatitis in patients with severe hypertriglyceridemia and cardiovascular disease in those with moderate hypertriglyceridemia. This review discusses the various genetic and acquired causes of hypertriglyceridemia, as well as current management strategies. Evidence supporting the different drug and non-drug approaches to treating hypertriglyceridemia is examined, and an easy to adopt step-by-step management strategy is presented.
Collapse
Affiliation(s)
- Vinaya Simha
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
15
|
Taghizadeh E, Mirzaei F, Jalilian N, Ghayour Mobarhan M, Ferns GA, Pasdar A. A novel mutation in
USF1
gene is associated with familial combined hyperlipidemia. IUBMB Life 2019; 72:616-623. [DOI: 10.1002/iub.2186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 10/01/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Eskandar Taghizadeh
- Department of Medical Genetics, Faculty of MedicineMashhad University of Medical Sciences Mashhad Iran
- Cellular and Molecular Research CenterYasuj University of Medical Sciences Yasuj Iran
| | - Farzaneh Mirzaei
- Department of Medical Genetics, Faculty of MedicineMashhad University of Medical Sciences Mashhad Iran
| | - Nazanin Jalilian
- Department of Clinical biochemistry, School of MedicineKermanshah University of Medical Sciences Kermanshah Iran
| | - Majid Ghayour Mobarhan
- Metabolic Syndrome Research Centre, School of MedicineMashhad University of Medical Sciences Mashhad Iran
| | - Gordon A. Ferns
- Department of Medical EducationBrighton and Sussex Medical School Perso Falmer Brighton UK
| | - Alireza Pasdar
- Department of Medical Genetics, Faculty of MedicineMashhad University of Medical Sciences Mashhad Iran
- Medical Genetics Research Centre, Faculty of MedicineMashhad University of Medical Sciences Mashhad Iran
- Division of Applied Medicine, Medical School, University of Aberdeen Foresterhill Aberdeen UK
| |
Collapse
|
16
|
Popa SC, Shin JA. The Intrinsically Disordered Loop in the USF1 bHLHZ Domain Modulates Its DNA-Binding Sequence Specificity in Hereditary Asthma. J Phys Chem B 2019; 123:9862-9871. [PMID: 31670516 DOI: 10.1021/acs.jpcb.9b06719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
USF1, a basic region/helix-loop-helix/leucine zipper (bHLHZ) transcription factor, binds to the E-box in the PAI-1 (plasminogen activator inhibitor) promoter. Two alleles containing the E-box control PAI-1 transcription; these alleles are termed "4G" and "5G" based on the G tract flanking E-box. USF1-governed transcription of PAI-1 is elevated in heritable asthma sufferers: the 4G/4G genotype has the highest plasma levels of PAI-1. While USF1 uses its basic region to bind E-box, we found that it uses its 12 amino-acid loop to recognize the flanking sequence and discern the single-nucleotide difference between the alleles. We used the bacterial one-hybrid and electrophoretic mobility shift assays to assess protein-DNA recognition, and circular dichroism to examine protein secondary structure. We mutated Ser233 and Thr234 in the USF1 bHLHZ loop to Ala to generate S233A and T234A. Interestingly, USF1 bHLHZ, S233A, and T234A prefer the 5G sequence (USF1 bHLHZ Kd values 4.1 ± 0.3 nM and 7.0 ± 0.4 nM for 5G and 4G, respectively), whereas studies in stimulated human mast cells showed a preference for 4G. We replaced the 8 amino-acid loop of transcription factor Max bHLHZ with the 12 amino-acid USF1 loop: this mutant now distinguishes the 4G/5G polymorphism-while Max bHLHZ does not-confirming that USF1 differentiation of the 4G/5G is driven by the loop.
Collapse
Affiliation(s)
- Serban C Popa
- Department of Chemistry , University of Toronto , 3359 Mississauga Road , Mississauga , Ontario L5L 1C6 , Canada
| | - Jumi A Shin
- Department of Chemistry , University of Toronto , 3359 Mississauga Road , Mississauga , Ontario L5L 1C6 , Canada
| |
Collapse
|
17
|
Castillejo-Lopez C, Pjanic M, Pirona AC, Hetty S, Wabitsch M, Wadelius C, Quertermous T, Arner E, Ingelsson E. Detailed Functional Characterization of a Waist-Hip Ratio Locus in 7p15.2 Defines an Enhancer Controlling Adipocyte Differentiation. iScience 2019; 20:42-59. [PMID: 31557715 PMCID: PMC6817687 DOI: 10.1016/j.isci.2019.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 07/10/2019] [Accepted: 09/05/2019] [Indexed: 12/22/2022] Open
Abstract
We combined CAGE sequencing in human adipocytes during differentiation with data from genome-wide association studies to identify an enhancer in the SNX10 locus on chromosome 7, presumably involved in body fat distribution. Using reporter assays and CRISPR-Cas9 gene editing in human cell lines, we characterized the role of the enhancer in adipogenesis. The enhancer was active during adipogenesis and responded strongly to insulin and isoprenaline. The allele associated with increased waist-hip ratio in human genetic studies was associated with higher enhancer activity. Mutations of the enhancer resulted in less adipocyte differentiation. RNA sequencing of cells with disrupted enhancer showed reduced expression of established adipocyte markers, such as ADIPOQ and LPL, and identified CHI3L1 on chromosome 1 as a potential gene involved in adipocyte differentiation. In conclusion, we identified and characterized an enhancer in the SNX10 locus and outlined its plausible mechanisms of action and downstream targets.
Collapse
Affiliation(s)
- Casimiro Castillejo-Lopez
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Milos Pjanic
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anna Chiara Pirona
- Department of Medical Sciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Susanne Hetty
- Department of Medical Sciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Martin Wabitsch
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Endocrinology and Diabetes, University of Ulm, Ulm, Germany
| | - Claes Wadelius
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Thomas Quertermous
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
| | - Erik Arner
- Laboratory for Applied Regulatory Genomics Network Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045 Japan
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medical Sciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden; Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
18
|
Sun Q, Li J, Li F, Li H, Bei S, Zhang X, Feng L. LncRNA LOXL1-AS1 facilitates the tumorigenesis and stemness of gastric carcinoma via regulation of miR-708-5p/USF1 pathway. Cell Prolif 2019; 52:e12687. [PMID: 31468594 PMCID: PMC6869681 DOI: 10.1111/cpr.12687] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/28/2019] [Accepted: 07/15/2019] [Indexed: 12/24/2022] Open
Abstract
Objectives As one of the most life‐threatening malignancies, gastric cancer is the third contributor of cancer mortalities globally. Increasing studies have proven the regulatory roles of lncRNAs in the development of diverse malignant tumours. But little is known about its function and molecular mechanism in gastric carcinoma. Materials and methods RT‐qPCR was performed to measure the expression pattern of LOXL1‐AS1 in gastric cancer. To ascertain its definite role, CCK‐8, EdU, Western blot, transwell and sphere formation assays were adopted. RNA pull‐down, RIP, ChIP and luciferase reporter assays were carried out to investigate the molecular mechanism of LOXL1‐AS1 in gastric carcinoma. Results LOXL1‐AS1 was highly expressed in tissues and cells of gastric cancer. The upregulation of LOXL1‐AS1 predicted poor prognosis in gastric carcinoma. Our findings demonstrated that LOXL1‐AS1 accelerated the deterioration of gastric cancer by inducing cell proliferation, migration, EMT and stemness. Moreover, the expression of USF1 in gastric cancer was higher than in normal control and LOXL1‐AS1 negatively modulated USF1. Functionally, LOXL1‐AS1 acted as a ceRNA to upregulate USF1 via sponging miR‐708‐5p. Besides, we confirmed USF1 promoted the transcription of stemness marker SOX2. Rescue experiments testified the stimulative role of LOXL1‐AS1/miR‐708‐5p/USF1 pathway in gastric cancer progression. It was also validated that LOXL1‐AS1 facilitated cell growth of gastric carcinoma in vivo. Conclusions Our study unravelled that LOXL1‐AS1/miR‐708‐5p/USF1 pathway contributed to the development of gastric cancer.
Collapse
Affiliation(s)
- Qi Sun
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Jian Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Fan Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Huanqin Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Songhua Bei
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaohong Zhang
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Li Feng
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Taghizadeh E, Esfehani RJ, Sahebkar A, Parizadeh SM, Rostami D, Mirinezhad M, Poursheikhani A, Mobarhan MG, Pasdar A. Familial combined hyperlipidemia: An overview of the underlying molecular mechanisms and therapeutic strategies. IUBMB Life 2019; 71:1221-1229. [PMID: 31271707 DOI: 10.1002/iub.2073] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 05/03/2019] [Indexed: 12/30/2022]
Abstract
Among different types of dyslipidemia, familial combined hyperlipidemia (FCHL) is the most common genetic disorder, which is characterized by at least two different forms of lipid abnormalities: hypercholesterolemia and hypertriglyceridemia. FCHL is an important cause of cardiovascular diseases. FCHL is a heterogeneous condition linked with some metabolic defects that are closely associated with FCHL. These metabolic features include dysfunctional adipose tissue, delayed clearance of triglyceride-rich lipoproteins, overproduction of very low-density lipoprotein and hepatic lipids, and defect in the clearance of low-density lipoprotein particles. There are also some genes associated with FCHL such as those affecting the metabolism and clearance of plasma lipoprotein particles. Due to the high prevalence of FCHL especially in cardiovascular patients, targeted treatment is ideal but this necessitates identification of the genetic background of patients. This review describes the metabolic pathways and associated genes that are implicated in FCHL pathogenesis. We also review existing and novel treatment options for FCHL. © 2019 IUBMB Life, 71(9):1221-1229, 2019.
Collapse
Affiliation(s)
- Eskandar Taghizadeh
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Reza Jafarzadeh Esfehani
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Medical Genetics Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mostafa Parizadeh
- Metabolic Syndrome Research Centre, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Daryoush Rostami
- Department of School Allied, Zabol University of Medical Sciences, Zabol, Iran
| | - Mohammadreza Mirinezhad
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arash Poursheikhani
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour Mobarhan
- Metabolic Syndrome Research Centre, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Pasdar
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Medical Genetics Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Division of Applied Medicine, Medical School, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
20
|
Faisal I, Cisneros-Montalvo S, Hamer G, Tuominen MM, Laurila PP, Tumiati M, Jauhiainen M, Kotaja N, Toppari J, Mäkelä JA, Kauppi L. Transcription Factor USF1 Is Required for Maintenance of Germline Stem Cells in Male Mice. Endocrinology 2019; 160:1119-1136. [PMID: 30759202 DOI: 10.1210/en.2018-01088] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/08/2019] [Indexed: 12/22/2022]
Abstract
A prerequisite for lifelong sperm production is that spermatogonial stem cells (SSCs) balance self-renewal and differentiation, yet factors required for this balance remain largely undefined. Using mouse genetics, we now demonstrate that the ubiquitously expressed transcription factor upstream stimulatory factor (USF)1 is critical for the maintenance of SSCs. We show that USF1 is not only detected in Sertoli cells as previously reported, but also in SSCs. Usf1-deficient mice display progressive spermatogenic decline as a result of age-dependent loss of SSCs. According to our data, the germ cell defect in Usf1-/- mice cannot be attributed to impairment of Sertoli cell development, maturation, or function, but instead is likely due to an inability of SSCs to maintain a quiescent state. SSCs of Usf1-/- mice undergo continuous proliferation, which provides an explanation for their age-dependent depletion. The proliferation-coupled exhaustion of SSCs in turn results in progressive degeneration of the seminiferous epithelium, gradual decrease in sperm production, and testicular atrophy. We conclude that the general transcription factor USF1 is indispensable for the proper maintenance of mammalian spermatogenesis.
Collapse
Affiliation(s)
- Imrul Faisal
- Genome-Scale Biology Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Doctoral Program in Biomedicine, Doctoral School in Health Sciences, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sheyla Cisneros-Montalvo
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
- Turku Doctoral Program of Molecular Medicine, University of Turku, Turku, Finland
| | - Geert Hamer
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Minna M Tuominen
- Genome-Scale Biology Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pirkka-Pekka Laurila
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Biomedicum, Helsinki, Finland
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Manuela Tumiati
- Genome-Scale Biology Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Matti Jauhiainen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Biomedicum, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Noora Kotaja
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Jorma Toppari
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Juho-Antti Mäkelä
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Liisa Kauppi
- Genome-Scale Biology Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
21
|
Malakar AK, Choudhury D, Halder B, Paul P, Uddin A, Chakraborty S. A review on coronary artery disease, its risk factors, and therapeutics. J Cell Physiol 2019; 234:16812-16823. [PMID: 30790284 DOI: 10.1002/jcp.28350] [Citation(s) in RCA: 558] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 12/19/2022]
Abstract
Coronary artery disease (CAD) is one of the major cardiovascular diseases affecting the global human population. This disease has been proved to be the major cause of death in both the developed and developing countries. Lifestyle, environmental factors, and genetic factors pose as risk factors for the development of cardiovascular disease. The prevalence of risk factors among healthy individuals elucidates the probable occurrence of CAD in near future. Genome-wide association studies have suggested the association of chromosome 9p21.3 in the premature onset of CAD. The risk factors of CAD include diabetes mellitus, hypertension, smoking, hyperlipidemia, obesity, homocystinuria, and psychosocial stress. The eradication and management of CAD has been established through extensive studies and trials. Antiplatelet agents, nitrates, β-blockers, calcium antagonists, and ranolazine are some of the few therapeutic agents used for the relief of symptomatic angina associated with CAD.
Collapse
Affiliation(s)
- Arup Kr Malakar
- Department of Biotechnology, Assam University, Silchar, Assam, India
| | | | - Binata Halder
- Department of Biotechnology, Assam University, Silchar, Assam, India
| | - Prosenjit Paul
- Department of Biotechnology, Assam University, Silchar, Assam, India
| | - Arif Uddin
- Department of Zoology, Moinul Hoque Choudhury Memorial Science College, Hailakandi, Assam, India
| | | |
Collapse
|
22
|
Chao HW, Chao SW, Lin H, Ku HC, Cheng CF. Homeostasis of Glucose and Lipid in Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2019; 20:298. [PMID: 30642126 PMCID: PMC6359196 DOI: 10.3390/ijms20020298] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 02/07/2023] Open
Abstract
Industrialized society-caused dysregular human behaviors and activities such as overworking, excessive dietary intake, and sleep deprivation lead to perturbations in the metabolism and the development of metabolic syndrome. Non-alcoholic fatty liver disease (NAFLD), the most common chronic liver disease worldwide, affects around 30% and 25% of people in Western and Asian countries, respectively, which leads to numerous medical costs annually. Insulin resistance is the major hallmark of NAFLD and is crucial in the pathogenesis and for the progression from NAFLD to non-alcoholic steatohepatitis (NASH). Excessive dietary intake of saturated fats and carbohydrate-enriched foods contributes to both insulin resistance and NAFLD. Once NAFLD is established, insulin resistance can promote the progression to the more severe state of liver endangerment like NASH. Here, we review current and potential studies for understanding the complexity between insulin-regulated glycolytic and lipogenic homeostasis and the underlying causes of NAFLD. We discuss how disruption of the insulin signal is associated with various metabolic disorders of glucoses and lipids that constitute both the metabolic syndrome and NAFLD.
Collapse
Affiliation(s)
- Hsu-Wen Chao
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Shi-Wei Chao
- Ph.D. Program in Biotechnology Research and Development, Taipei Medical University, Taipei 11031, Taiwan.
| | - Heng Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Ph.D. Program in Biotechnology Research and Development, Taipei Medical University, Taipei 11031, Taiwan.
| | - Hui-Chen Ku
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan.
| | - Ching-Feng Cheng
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan.
- Department of Pediatrics, Tzu Chi University, Hualien 97004, Taiwan.
| |
Collapse
|
23
|
The Regulation of Cbf1 by PAS Kinase Is a Pivotal Control Point for Lipogenesis vs. Respiration in Saccharomyces cerevisiae. G3-GENES GENOMES GENETICS 2019; 9:33-46. [PMID: 30381292 PMCID: PMC6325914 DOI: 10.1534/g3.118.200663] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PAS kinase 1 (Psk1) is a key regulator of respiration in Saccharomyces cerevisiae. Herein the molecular mechanisms of this regulation are explored through the characterization of its substrate, Centromere binding factor 1 (Cbf1). CBF1-deficient yeast displayed a significant decrease in cellular respiration, while PAS kinase-deficient yeast, or yeast harboring a Cbf1 phosphosite mutant (T211A) displayed a significant increase. Transmission electron micrographs showed an increased number of mitochondria in PAS kinase-deficient yeast consistent with the increase in respiration. Although the CBF1-deficient yeast did not appear to have an altered number of mitochondria, a mitochondrial proteomics study revealed significant differences in the mitochondrial composition of CBF1-deficient yeast including altered Atp3 levels, a subunit of the mitochondrial F1-ATP synthase complex. Both beta-galactosidase reporter assays and western blot analysis confirmed direct transcriptional control of ATP3 by Cbf1. In addition, we confirmed the regulation of yeast lipid genes LAC1 and LAG1 by Cbf1. The human homolog of Cbf1, Upstream transcription factor 1 (USF1), is also known to be involved in lipid biogenesis. Herein, we provide the first evidence for a role of USF1 in respiration since it appeared to complement Cbf1in vivo as determined by respiration phenotypes. In addition, we confirmed USF1 as a substrate of human PAS kinase (hPASK) in vitro. Combined, our data supports a model in which Cbf1/USF1 functions to partition glucose toward respiration and away from lipid biogenesis, while PAS kinase inhibits respiration in part through the inhibition of Cbf1/USF1.
Collapse
|
24
|
Ruuth M, Soronen J, Kaiharju E, Merikanto K, Perttilä J, Metso J, Lee-Rueckert M, Taskinen MR, Kovanen PT, Öörni K, Olkkonen VM, Jauhiainen MS, Laurila PP. USF1 deficiency alleviates inflammation, enhances cholesterol efflux and prevents cholesterol accumulation in macrophages. Lipids Health Dis 2018; 17:285. [PMID: 30545366 PMCID: PMC6293625 DOI: 10.1186/s12944-018-0930-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/26/2018] [Indexed: 12/23/2022] Open
Abstract
Background The focus of studies on high-density lipoproteins (HDL) has shifted from HDL-cholesterol (HDL-C) to HDL function. We recently demonstrated that low USF1 expression in mice and humans associates with high plasma HDL-C and low triglyceride levels, as well as protection against obesity, insulin resistance, and atherosclerosis. Here, we studied the impact of USF1 deficiency on HDL functional capacity and macrophage atherogenic functions, including inflammation, cholesterol efflux, and cholesterol accumulation. Methods We used a congenic Usf1 deficient mice in C57Bl/6JRccHsd background and blood samples were collected to isolate HDL for structural and functional studies. Lentiviral preparations containing the USF1 silencing shRNA expression vector were used to silence USF1 in human THP-1 and Huh-7 cells. Cholesterol efflux from acetyl-LDL loaded THP-1 macrophages was measured using HDL and plasma as acceptors. Gene expression analysis from USF1 silenced peritoneal macrophages was carried out using Affymetrix protocols. Results We show that Usf1 deficiency not only increases HDL-C levels in vivo, consistent with elevated ABCA1 protein expression in hepatic cell lines, but also improves the functional capacity of HDL particles. HDL particles derived from Usf1 deficient mice remove cholesterol more efficiently from macrophages, attributed to their higher contents of phospholipids. Furthermore, silencing of USF1 in macrophages enhanced the cholesterol efflux capacity of these cells. These findings are consistent with reduced inflammatory burden of USF1 deficient macrophages, manifested by reduced secretion of pro-inflammatory cytokines MCP-1 and IL-1β and protection against inflammation-induced macrophage cholesterol accumulation in a cell-autonomous manner. Conclusions Our findings identify USF1 as a novel factor regulating HDL functionality, showing that USF1 inactivation boosts cholesterol efflux, reduces macrophage inflammation and attenuates macrophage cholesterol accumulation, linking improved macrophage cholesterol metabolism and inflammatory pathways to the antiatherogenic function of USF1 deficiency.
Collapse
Affiliation(s)
- Maija Ruuth
- Wihuri Research Institute, FI-00290, Helsinki, Finland.,Research Program Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Jarkko Soronen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, FI-00251, Helsinki, Finland.,Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland
| | - Essi Kaiharju
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, FI-00251, Helsinki, Finland
| | - Krista Merikanto
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, FI-00251, Helsinki, Finland
| | - Julia Perttilä
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland
| | - Jari Metso
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, FI-00251, Helsinki, Finland
| | | | - Marja-Riitta Taskinen
- Diabetes and Obesity Research Program, University of Helsinki, FI-00014, Helsinki, Finland
| | | | | | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland.,Department of Anatomy, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland
| | - Matti S Jauhiainen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, FI-00251, Helsinki, Finland. .,Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290, Helsinki, Finland.
| | - Pirkka-Pekka Laurila
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, FI-00251, Helsinki, Finland.,Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland.,Institute for Molecular Medicine Finland, FIMM, FI-00251, Helsinki, Finland
| |
Collapse
|
25
|
Wang W, Yao S, Jiang H, Dong J, Cui X, Tian X, Guo Y, Zhang S. Upstream transcription factor 1 prompts malignancies of cervical cancer primarily by transcriptionally activating p65 expression. Exp Ther Med 2018; 16:4415-4422. [PMID: 30542391 PMCID: PMC6257725 DOI: 10.3892/etm.2018.6758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 07/20/2017] [Indexed: 11/21/2022] Open
Abstract
Cervical cancer is the third-most common cause of female cancer-related mortality worldwide. In cervical cancer, aberrant activation of nuclear factor (NF)-κB signaling is widely reported. However, the transcriptional regulation of NF-κB signaling remains unclear. The present study aimed to explore the underlying mechanism in which NF-κB signaling was activated in cervical cancer cells. Initially, the expression of p65 was demonstrated to be markedly enhanced in grade II, III or IV cervical cancer tissues compared with that of normal cervical tissues, indicating that p65 expression was correlated with tumor grade. In HeLa and CaSki cells, overexpression of p65 markedly enhanced cervical cancer cell invasion and migration. Further experiments demonstrated that p65 overexpression significantly increased the phosphorylation levels of protein kinase B (AKT) and p38. Dual luciferase reporter and chromatin immunoprecipitation assays demonstrated that USF1 was able to bind the promoter region of p65, thereby enhancing the transcriptional activation of p65. Notably, when p65 was silenced, the phosphorylation levels of AKT and p38 were suppressed even in cells transfected with adenovirus vectors expressing upstream transcription factor 1 (USF1). These data indicated that USF1 prompted cervical cancer progression primarily by transcriptionally activating p65. In conclusion, the present study demonstrated that USF1 was able to activate the transcription of p65, thereby enhancing the malignancy of cervical cancer cells.
Collapse
Affiliation(s)
- Wen Wang
- Department of Obstetrics and Gynecology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shujuan Yao
- Department of Obstetrics and Gynecology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Hongjing Jiang
- Department of Obstetrics and Gynecology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Jing Dong
- Department of Obstetrics and Gynecology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Xiujuan Cui
- Department of Obstetrics and Gynecology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Xiangyu Tian
- Department of Medical Imaging, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Yanyan Guo
- Department of Obstetrics and Gynecology, Shandong Police Hospital, Jinan, Shandong 250001, P.R. China
| | - Shiqian Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
26
|
Wang T, Yao W, Shao Y, Zheng R, Huang F. PCAF fine-tunes hepatic metabolic syndrome, inflammatory disease, and cancer. J Cell Mol Med 2018; 22:5787-5800. [PMID: 30216660 PMCID: PMC6237576 DOI: 10.1111/jcmm.13877] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/07/2018] [Indexed: 02/07/2023] Open
Abstract
The P300/CBP‐associating factor (PCAF), a histone acetyltransferase, is involved in metabolic and pathogenic diseases, particularly of the liver. The effects of PCAF on fine‐tuning liver diseases are extremely complex and vary according to different pathological conditions. This enzyme has dichotomous functions, depending on differently modified sites, which regulate the activities of various enzymes, metabolic functions, and gene expression. Here, we summarize the most recent findings on the functions and targets of PCAF in various metabolic and immunological processes in the liver and review these new discoveries and models of PCAF biology in three areas: hepatic metabolic syndrome, inflammatory disease, and cancer. Finally, we discuss the potential implications of these findings for therapeutic interventions in liver diseases.
Collapse
Affiliation(s)
- Tongxin Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Weilei Yao
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yafei Shao
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ruilong Zheng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Feiruo Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
27
|
Ozsait‐Selcuk B, Komurcu‐Bayrak E, Jylhä M, Luukkaala T, Perola M, Kristiansson K, Mononen N, Hurme M, Kähönen M, Goebeler S, Laaksonen R, Hervonen A, Erginel‐Unaltuna N, Karhunen P, Lehtimäki T. The
rs2516839
variation of
USF1
gene is associated with 4‐year mortality of nonagenarian women: The Vitality 90+ study. Ann Hum Genet 2018; 83:34-45. [DOI: 10.1111/ahg.12282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/17/2018] [Accepted: 07/30/2018] [Indexed: 11/29/2022]
Affiliation(s)
- B. Ozsait‐Selcuk
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center ‐ Tampere, Faculty of Medicine and Life Sciences University of Tampere Tampere Finland
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine Istanbul University Istanbul Turkey
| | - E. Komurcu‐Bayrak
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center ‐ Tampere, Faculty of Medicine and Life Sciences University of Tampere Tampere Finland
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine Istanbul University Istanbul Turkey
| | - M. Jylhä
- Gerontology Research Center (GEREC), University of Tampere; School of Health Sciences University of Tampere Tampere Finland
| | - T. Luukkaala
- Tampere School of Health Sciences, University of Tampere, Tampere; Science Center Pirkanmaa Hospital District Finland
| | - M. Perola
- Department of Health National Institute for Health and Welfare Helsinki Finland
| | - K. Kristiansson
- Department of Microbiology and Immunology, Faculty of Medicine and Life Sciences University of Tampere Tampere Finland
| | - N. Mononen
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center ‐ Tampere, Faculty of Medicine and Life Sciences University of Tampere Tampere Finland
| | - M. Hurme
- Department of Microbiology and Immunology, Faculty of Medicine and Life Sciences University of Tampere Tampere Finland
| | - M. Kähönen
- Department of Clinical Physiology, Tampere University Hospital, and Finnish Cardiovascular Research Center ‐ Tampere, Faculty of Medicine and Life Sciences University of Tampere Tampere Finland
| | - S. Goebeler
- Department of Forensic Medicine, University of Tampere, Fimlab Laboratories Pirkanmaa Hospital District Tampere Finland
| | - R. Laaksonen
- Medical School, University of Tampere; Finnish Clinical Biobank University Hospital of Tampere Tampere Finland
| | - A. Hervonen
- Gerontology Research Center (GEREC), University of Tampere; School of Health Sciences University of Tampere Tampere Finland
| | - N. Erginel‐Unaltuna
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine Istanbul University Istanbul Turkey
| | - P.J. Karhunen
- Department of Clinical Chemistry, Fimlab Laboratories, and Department of Forensic Medicine, Finnish Cardiovascular Research Center ‐ Tampere, Faculty of Medicine and Life Sciences University of Tampere Tampere Finland
| | - T. Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center ‐ Tampere, Faculty of Medicine and Life Sciences University of Tampere Tampere Finland
| |
Collapse
|
28
|
A novel homozygous mutation in the glycerol-3-phosphate dehydrogenase 1 gene in a Chinese patient with transient infantile hypertriglyceridemia: a case report. BMC Gastroenterol 2018; 18:96. [PMID: 29940878 PMCID: PMC6020200 DOI: 10.1186/s12876-018-0827-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 06/19/2018] [Indexed: 11/10/2022] Open
Abstract
Background Transient infantile hypertriglyceridemia (HTGTI) is an autosomal recessive disorder caused by mutations in the glycerol-3-phosphate dehydrogenase 1 (GPD1) gene. Case presentation We report a case of HTGTI in a Chinese female infant. She presented with hepatomegaly, hypertriglyceridemia, moderately elevated transaminases, and hepatic steatosis at 3.5 months of age. A novel mutation c.523C>T, p. (Q175*) was identified in GPD1. The patient was a homozygote and her parents were heterozygous for the mutation. Ultrastructural study showed intrahepatocytic lipid droplets. Conclusions This is the first reported case of HTGTI in Chinese, expanding the worldwide distribution of HTGTI and the mutation spectrum of GPD1. Electronic supplementary material The online version of this article (10.1186/s12876-018-0827-6) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Marnetto D, Mantica F, Molineris I, Grassi E, Pesando I, Provero P. Evolutionary Rewiring of Human Regulatory Networks by Waves of Genome Expansion. Am J Hum Genet 2018; 102:207-218. [PMID: 29357977 DOI: 10.1016/j.ajhg.2017.12.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 12/15/2017] [Indexed: 01/09/2023] Open
Abstract
Genome expansion is believed to be an important driver of the evolution of gene regulation. To investigate the role of a newly arising sequence in rewiring regulatory networks, we estimated the age of each region of the human genome by applying maximum parsimony to genome-wide alignments with 100 vertebrates. We then studied the age distribution of several types of functional regions, with a focus on regulatory elements. The age distribution of regulatory elements reveals the extensive use of newly formed genomic sequence in the evolution of regulatory interactions. Many transcription factors have expanded their repertoire of targets through waves of genomic expansions that can be traced to specific evolutionary times. Repeated elements contributed a major part of such expansion: many classes of such elements are enriched in binding sites of one or a few specific transcription factors, whose binding sites are localized in specific portions of the element and characterized by distinctive motif words. These features suggest that the binding sites were available as soon as the new sequence entered the genome, rather than being created later by accumulation of point mutations. By comparing the age of regulatory regions to the evolutionary shift in expression of nearby genes, we show that rewiring through genome expansion played an important role in shaping human regulatory networks.
Collapse
|
30
|
Li YF, Altman RB. Systematic target function annotation of human transcription factors. BMC Biol 2018; 16:4. [PMID: 29325558 PMCID: PMC5795274 DOI: 10.1186/s12915-017-0469-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 12/06/2017] [Indexed: 01/03/2023] Open
Abstract
Background Transcription factors (TFs), the key players in transcriptional regulation, have attracted great experimental attention, yet the functions of most human TFs remain poorly understood. Recent capabilities in genome-wide protein binding profiling have stimulated systematic studies of the hierarchical organization of human gene regulatory network and DNA-binding specificity of TFs, shedding light on combinatorial gene regulation. We show here that these data also enable a systematic annotation of the biological functions and functional diversity of TFs. Result We compiled a human gene regulatory network for 384 TFs covering the 146,096 TF–target gene (TF–TG) relationships, extracted from over 850 ChIP-seq experiments as well as the literature. By integrating this network of TF–TF and TF–TG relationships with 3715 functional concepts from six sources of gene function annotations, we obtained over 9000 confident functional annotations for 279 TFs. We observe extensive connectivity between TFs and Mendelian diseases, GWAS phenotypes, and pharmacogenetic pathways. Further, we show that TFs link apparently unrelated functions, even when the two functions do not share common genes. Finally, we analyze the pleiotropic functions of TFs and suggest that the increased number of upstream regulators contributes to the functional pleiotropy of TFs. Conclusion Our computational approach is complementary to focused experimental studies on TF functions, and the resulting knowledge can guide experimental design for the discovery of unknown roles of TFs in human disease and drug response. Electronic supplementary material The online version of this article (doi:10.1186/s12915-017-0469-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yong Fuga Li
- Stanford Genome Technology Center, Stanford, CA, USA. .,Department of Bioengineering, Stanford University, Stanford, CA, USA. .,Present address: Department of Bioinformatics, Illumina Inc., San Diego, CA, USA.
| | - Russ B Altman
- Department of Bioengineering, Stanford University, Stanford, CA, USA. .,Department of Genetics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
31
|
Jalili V, Matteucci M, Masseroli M, Ceri S. Explorative visual analytics on interval-based genomic data and their metadata. BMC Bioinformatics 2017; 18:536. [PMID: 29202689 PMCID: PMC5715631 DOI: 10.1186/s12859-017-1945-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 11/19/2017] [Indexed: 02/07/2023] Open
Abstract
Background With the wide-spreading of public repositories of NGS processed data, the availability of user-friendly and effective tools for data exploration, analysis and visualization is becoming very relevant. These tools enable interactive analytics, an exploratory approach for the seamless “sense-making” of data through on-the-fly integration of analysis and visualization phases, suggested not only for evaluating processing results, but also for designing and adapting NGS data analysis pipelines. Results This paper presents abstractions for supporting the early analysis of NGS processed data and their implementation in an associated tool, named GenoMetric Space Explorer (GeMSE). This tool serves the needs of the GenoMetric Query Language, an innovative cloud-based system for computing complex queries over heterogeneous processed data. It can also be used starting from any text files in standard BED, BroadPeak, NarrowPeak, GTF, or general tab-delimited format, containing numerical features of genomic regions; metadata can be provided as text files in tab-delimited attribute-value format. GeMSE allows interactive analytics, consisting of on-the-fly cycling among steps of data exploration, analysis and visualization that help biologists and bioinformaticians in making sense of heterogeneous genomic datasets. By means of an explorative interaction support, users can trace past activities and quickly recover their results, seamlessly going backward and forward in the analysis steps and comparative visualizations of heatmaps. Conclusions GeMSE effective application and practical usefulness is demonstrated through significant use cases of biological interest. GeMSE is available at http://www.bioinformatics.deib.polimi.it/GeMSE/, and its source code is available at https://github.com/Genometric/GeMSE under GPLv3 open-source license.
Collapse
Affiliation(s)
- Vahid Jalili
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano, 20133, Italy.
| | - Matteo Matteucci
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano, 20133, Italy
| | - Marco Masseroli
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano, 20133, Italy
| | - Stefano Ceri
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano, 20133, Italy
| |
Collapse
|
32
|
Ellis KL, Pang J, Chan DC, Hooper AJ, Bell DA, Burnett JR, Watts GF. Familial combined hyperlipidemia and hyperlipoprotein(a) as phenotypic mimics of familial hypercholesterolemia: Frequencies, associations and predictions. J Clin Lipidol 2016; 10:1329-1337.e3. [DOI: 10.1016/j.jacl.2016.08.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/11/2016] [Accepted: 08/16/2016] [Indexed: 12/11/2022]
|
33
|
Laurila PP, Soronen J, Kooijman S, Forsström S, Boon MR, Surakka I, Kaiharju E, Coomans CP, Van Den Berg SAA, Autio A, Sarin AP, Kettunen J, Tikkanen E, Manninen T, Metso J, Silvennoinen R, Merikanto K, Ruuth M, Perttilä J, Mäkelä A, Isomi A, Tuomainen AM, Tikka A, Ramadan UA, Seppälä I, Lehtimäki T, Eriksson J, Havulinna A, Jula A, Karhunen PJ, Salomaa V, Perola M, Ehnholm C, Lee-Rueckert M, Van Eck M, Roivainen A, Taskinen MR, Peltonen L, Mervaala E, Jalanko A, Hohtola E, Olkkonen VM, Ripatti S, Kovanen PT, Rensen PCN, Suomalainen A, Jauhiainen M. USF1 deficiency activates brown adipose tissue and improves cardiometabolic health. Sci Transl Med 2016; 8:323ra13. [PMID: 26819196 DOI: 10.1126/scitranslmed.aad0015] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
USF1 (upstream stimulatory factor 1) is a transcription factor associated with familial combined hyperlipidemia and coronary artery disease in humans. However, whether USF1 is beneficial or detrimental to cardiometabolic health has not been addressed. By inactivating USF1 in mice, we demonstrate protection against diet-induced dyslipidemia, obesity, insulin resistance, hepatic steatosis, and atherosclerosis. The favorable plasma lipid profile, including increased high-density lipoprotein cholesterol and decreased triglycerides, was coupled with increased energy expenditure due to activation of brown adipose tissue (BAT). Usf1 inactivation directs triglycerides from the circulation to BAT for combustion via a lipoprotein lipase-dependent mechanism, thus enhancing plasma triglyceride clearance. Mice lacking Usf1 displayed increased BAT-facilitated, diet-induced thermogenesis with up-regulation of mitochondrial respiratory chain complexes, as well as increased BAT activity even at thermoneutrality and after BAT sympathectomy. A direct effect of USF1 on BAT activation was demonstrated by an amplified adrenergic response in brown adipocytes after Usf1 silencing, and by augmented norepinephrine-induced thermogenesis in mice lacking Usf1. In humans, individuals carrying SNP (single-nucleotide polymorphism) alleles that reduced USF1 mRNA expression also displayed a beneficial cardiometabolic profile, featuring improved insulin sensitivity, a favorable lipid profile, and reduced atherosclerosis. Our findings identify a new molecular link between lipid metabolism and energy expenditure, and point to the potential of USF1 as a therapeutic target for cardiometabolic disease.
Collapse
Affiliation(s)
- Pirkka-Pekka Laurila
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland. Department of Medical Genetics, University of Helsinki, Helsinki FI-00014, Finland. Institute for Molecular Medicine Finland, FIMM, Helsinki FI-00251, Finland.
| | - Jarkko Soronen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland. Institute for Molecular Medicine Finland, FIMM, Helsinki FI-00251, Finland. Minerva Foundation Institute for Medical Research, Helsinki FI-00290, Finland
| | - Sander Kooijman
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden 2333 ZA, Netherlands. Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, Netherlands
| | - Saara Forsström
- Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki FI-00014, Finland
| | - Mariëtte R Boon
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden 2333 ZA, Netherlands. Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, Netherlands
| | - Ida Surakka
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland. Institute for Molecular Medicine Finland, FIMM, Helsinki FI-00251, Finland
| | - Essi Kaiharju
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland
| | - Claudia P Coomans
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden 2333 ZA, Netherlands. Department of Molecular Cell Biology, Leiden University Medical Center, Leiden 2333 ZA, Netherlands
| | | | - Anu Autio
- Turku PET Centre, University of Turku and Turku University Hospital, Turku FI-20520, Finland
| | - Antti-Pekka Sarin
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland. Institute for Molecular Medicine Finland, FIMM, Helsinki FI-00251, Finland
| | - Johannes Kettunen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland. Institute for Molecular Medicine Finland, FIMM, Helsinki FI-00251, Finland. Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu FI-90014, Finland
| | - Emmi Tikkanen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland. Institute for Molecular Medicine Finland, FIMM, Helsinki FI-00251, Finland. Hjelt Institute, University of Helsinki, Helsinki FI-00014, Finland
| | - Tuula Manninen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland. Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki FI-00014, Finland
| | - Jari Metso
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland
| | | | - Krista Merikanto
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland
| | - Maija Ruuth
- Wihuri Research Institute, Helsinki FI-00290, Finland
| | - Julia Perttilä
- Minerva Foundation Institute for Medical Research, Helsinki FI-00290, Finland
| | - Anne Mäkelä
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu FI-90014, Finland
| | - Ayaka Isomi
- Hiroshima University, Hiroshima 730-0053, Japan
| | - Anita M Tuomainen
- Institute of Dentistry, University of Helsinki, Helsinki FI-00014, Finland
| | - Anna Tikka
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland
| | - Usama Abo Ramadan
- Experimental MRI Laboratory, Department of Neurology, Helsinki University Central Hospital, Helsinki FI-00290, Finland
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories, and Tampere University School of Medicine, Tampere FI-33014, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Tampere University School of Medicine, Tampere FI-33014, Finland
| | - Johan Eriksson
- Department of Health, National Institute for Health and Welfare, Helsinki FI-00271, Finland. Folkhälsan Research Centre, Helsinki FI-00251, Finland. Unit of General Practice, Helsinki University Central Hospital, Helsinki FI-00290, Finland. Department of General Practice and Primary Health Care, University of Helsinki, Helsinki FI-00014, Finland
| | - Aki Havulinna
- Department of Health, National Institute for Health and Welfare, Helsinki FI-00271, Finland
| | - Antti Jula
- Department of Health, National Institute for Health and Welfare, Helsinki FI-00271, Finland
| | - Pekka J Karhunen
- Department of Clinical Chemistry, Fimlab Laboratories, and Tampere University School of Medicine, Tampere FI-33014, Finland
| | - Veikko Salomaa
- Department of Health, National Institute for Health and Welfare, Helsinki FI-00271, Finland
| | - Markus Perola
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland
| | - Christian Ehnholm
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland
| | | | - Miranda Van Eck
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden 2333 ZA, Netherlands
| | - Anne Roivainen
- Turku PET Centre, University of Turku and Turku University Hospital, Turku FI-20520, Finland. Turku Center for Disease Modeling, University of Turku, Turku FI-20520, Finland
| | - Marja-Riitta Taskinen
- Diabetes and Obesity Research Program, University of Helsinki, Helsinki FI-00014, Finland
| | | | - Eero Mervaala
- Institute of Biomedicine, University of Helsinki, Helsinki FI-00014, Finland
| | - Anu Jalanko
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland
| | - Esa Hohtola
- Department of Genetics and Physiology, University of Oulu, Oulu FI-90014, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Helsinki FI-00290, Finland
| | - Samuli Ripatti
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland. Institute for Molecular Medicine Finland, FIMM, Helsinki FI-00251, Finland. Hjelt Institute, University of Helsinki, Helsinki FI-00014, Finland. Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | | | - Patrick C N Rensen
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden 2333 ZA, Netherlands. Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden 2333 ZA, Netherlands
| | - Anu Suomalainen
- Molecular Neurology, Research Programs Unit, University of Helsinki, Helsinki FI-00014, Finland. Department of Neurology, Helsinki University Central Hospital, Helsinki FI-00290, Finland. Neuroscience Center, University of Helsinki, Helsinki FI-00014, Finland
| | - Matti Jauhiainen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Helsinki FI-00251, Finland.
| |
Collapse
|
34
|
Sayols-Baixeras S, Subirana I, Lluis-Ganella C, Civeira F, Roquer J, Do AN, Absher D, Cenarro A, Muñoz D, Soriano-Tárraga C, Jiménez-Conde J, Ordovas JM, Senti M, Aslibekyan S, Marrugat J, Arnett DK, Elosua R. Identification and validation of seven new loci showing differential DNA methylation related to serum lipid profile: an epigenome-wide approach. The REGICOR study. Hum Mol Genet 2016; 25:4556-4565. [PMID: 28173150 PMCID: PMC6284258 DOI: 10.1093/hmg/ddw285] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 07/13/2016] [Accepted: 08/19/2016] [Indexed: 11/13/2022] Open
Abstract
Lipid traits (total, low-density and high-density lipoprotein cholesterol, and triglycerides) are risk factors for cardiovascular disease. DNA methylation is not only an inherited but also modifiable epigenetic mark that has been related to cardiovascular risk factors. Our aim was to identify loci showing differential DNA methylation related to serum lipid levels. Blood DNA methylation was assessed using the Illumina Human Methylation 450 BeadChip. A two-stage epigenome-wide association study was performed, with a discovery sample in the REGICOR study (n = 645) and validation in the Framingham Offspring Study (n = 2,542). Fourteen CpG sites located in nine genes (SREBF1, SREBF2, PHOSPHO1, SYNGAP1, ABCG1, CPT1A, MYLIP, TXNIP and SLC7A11) and 2 intergenic regions showed differential methylation in association with lipid traits. Six of these genes and 1 intergenic region were new discoveries showing differential methylation related to total cholesterol (SREBF2), HDL-cholesterol (PHOSPHO1, SYNGAP1 and an intergenic region in chromosome 2) and triglycerides (MYLIP, TXNIP and SLC7A11). These CpGs explained 0.7%, 9.5% and 18.9% of the variability of total cholesterol, HDL cholesterol and triglycerides in the Framingham Offspring Study, respectively. The expression of the genes SREBF2 and SREBF1 was inversely associated with methylation of their corresponding CpGs (P-value = 0.0042 and 0.0045, respectively) in participants of the GOLDN study (n = 98). In turn, SREBF1 expression was directly associated with HDL cholesterol (P-value = 0.0429). Genetic variants in SREBF1, PHOSPHO1, ABCG1 and CPT1A were also associated with lipid profile. Further research is warranted to functionally validate these new loci and assess the causality of new and established associations between these differentially methylated loci and lipid metabolism.
Collapse
Affiliation(s)
- S Sayols-Baixeras
- Cardiovascular Epidemiology and Genetics Research Group, IMIM (Hospital
del Mar Medical Research Institute), Barcelona, Catalonia, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| | - I Subirana
- Cardiovascular Epidemiology and Genetics Research Group, IMIM (Hospital
del Mar Medical Research Institute), Barcelona, Catalonia, Spain
- CIBER Epidemiology and Public Health, Barcelona, Catalonia, Spain
| | - C Lluis-Ganella
- Cardiovascular Epidemiology and Genetics Research Group, IMIM (Hospital
del Mar Medical Research Institute), Barcelona, Catalonia, Spain
| | - F Civeira
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis,
Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza,
Spain
| | - J Roquer
- Department of Neurology, Neurovascular Research Group, IMIM (Hospital
del Mar Medical Research Institute), Barcelona 08003, Catalonia, Spain
| | - AN Do
- Department of Epidemiology, University of Alabama at Birmingham,
Birmingham, AL, USA
| | - D Absher
- Hudson Alpha Institute for Biotechnology, Huntsville, AL, USA
| | - A Cenarro
- Unidad Clínica y de Investigación en Lípidos y Arteriosclerosis,
Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria Aragón, Zaragoza,
Spain
| | - D Muñoz
- Cardiovascular Risk and Nutrition Research Group, IMIM (Hospital del Mar
Medical Research Institute), Barcelona, Catalonia, Spain
| | - C Soriano-Tárraga
- Department of Neurology, Neurovascular Research Group, IMIM (Hospital
del Mar Medical Research Institute), Barcelona 08003, Catalonia, Spain
| | - J Jiménez-Conde
- Department of Neurology, Neurovascular Research Group, IMIM (Hospital
del Mar Medical Research Institute), Barcelona 08003, Catalonia, Spain
| | - J M Ordovas
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts
University, Boston, MA, USA
| | - M Senti
- Department of Experimental and Health Sciences, Pompeu Fabra
University, Barcelona, Catalonia, Spain
| | - S Aslibekyan
- Department of Epidemiology, University of Alabama at Birmingham,
Birmingham, AL, USA
| | - J Marrugat
- Cardiovascular Epidemiology and Genetics Research Group, IMIM (Hospital
del Mar Medical Research Institute), Barcelona, Catalonia, Spain
| | - D K Arnett
- Dean's Office, College of Public Health, University of Kentucky,
Lexington, KY, USA
| | - R Elosua
- Cardiovascular Epidemiology and Genetics Research Group, IMIM (Hospital
del Mar Medical Research Institute), Barcelona, Catalonia, Spain
| |
Collapse
|
35
|
Diamanti K, Umer HM, Kruczyk M, Dąbrowski MJ, Cavalli M, Wadelius C, Komorowski J. Maps of context-dependent putative regulatory regions and genomic signal interactions. Nucleic Acids Res 2016; 44:9110-9120. [PMID: 27625394 PMCID: PMC5100580 DOI: 10.1093/nar/gkw800] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 08/31/2016] [Indexed: 12/24/2022] Open
Abstract
Gene transcription is regulated mainly by transcription factors (TFs). ENCODE and Roadmap Epigenomics provide global binding profiles of TFs, which can be used to identify regulatory regions. To this end we implemented a method to systematically construct cell-type and species-specific maps of regulatory regions and TF-TF interactions. We illustrated the approach by developing maps for five human cell-lines and two other species. We detected ∼144k putative regulatory regions among the human cell-lines, with the majority of them being ∼300 bp. We found ∼20k putative regulatory elements in the ENCODE heterochromatic domains suggesting a large regulatory potential in the regions presumed transcriptionally silent. Among the most significant TF interactions identified in the heterochromatic regions were CTCF and the cohesin complex, which is in agreement with previous reports. Finally, we investigated the enrichment of the obtained putative regulatory regions in the 3D chromatin domains. More than 90% of the regions were discovered in the 3D contacting domains. We found a significant enrichment of GWAS SNPs in the putative regulatory regions. These significant enrichments provide evidence that the regulatory regions play a crucial role in the genomic structural stability. Additionally, we generated maps of putative regulatory regions for prostate and colorectal cancer human cell-lines.
Collapse
Affiliation(s)
- Klev Diamanti
- Department of Cell and Molecular Biology, Uppsala University, Uppsala SE-751-24, Sweden
| | - Husen M Umer
- Department of Cell and Molecular Biology, Uppsala University, Uppsala SE-751-24, Sweden
| | - Marcin Kruczyk
- Department of Cell and Molecular Biology, Uppsala University, Uppsala SE-751-24, Sweden
| | - Michał J Dąbrowski
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala SE-751-08, Sweden
| | - Marco Cavalli
- Institute of Computer Science, Polish Academy of Sciences, Warsaw 012-48, Poland
| | - Claes Wadelius
- Institute of Computer Science, Polish Academy of Sciences, Warsaw 012-48, Poland
| | - Jan Komorowski
- Department of Cell and Molecular Biology, Uppsala University, Uppsala SE-751-24, Sweden .,Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala SE-751-08, Sweden
| |
Collapse
|
36
|
Zhang MC, Shen Y, Xue A, He M, Cresswell N, Li L, Zhao Z, Jiang Y, Burke A. Morphologic features of culprit lesions in sudden coronary death with family history of premature coronary artery disease. Forensic Sci Int 2016; 266:412-415. [PMID: 27419614 DOI: 10.1016/j.forsciint.2016.06.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 06/23/2016] [Accepted: 06/26/2016] [Indexed: 11/25/2022]
Abstract
The morphologic features of familial coronary artery disease (CAD) resulting in sudden coronary death (SCD) are poorly studied. The presence and type of culprit lesions may have important implications in the genetic basis for familial heart disease. Autopsies of SCD victims over a 5-year period from a statewide medical examiner's office were studied. Premature familial disease was defined as sudden death at ≤50 years in women and ≤45 years in men, with premature SCD or acute coronary syndrome in a first-degree relative. Culprit lesion was defined as acute plaque rupture, plaque erosion, and severe narrowing without thrombus (stable plaque). There were 174 acute plaque ruptures (age 49±10 years, 9% women), 49 plaque erosions (age 45±8 years, 37% women), and 213 stable plaques (age 53±11 years, 22% women). There were 8 plaque rupture with family history. There were 9 plaque erosions with family history. There were 7 stable plaques with family history. The rate of familial history in premature coronary disease was 18.4% in erosions, 4.6% in ruptures (p=.02 vs. erosion), and 3.3% in stable plaque (p=.002 vs. erosion). We concluded that the frequency of family history of premature sudden death due to CAD may be higher in plaque erosion as compared to patients dying with acute plaque rupture or stable plaque.
Collapse
Affiliation(s)
- Ming Chang Zhang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yiwen Shen
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Aimin Xue
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Meng He
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Nathaniel Cresswell
- Department of Pathology, Georgetown University Hospital, Washington, DC 20007, USA
| | - Ling Li
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; University of Maryland Medical System, Baltimore, MD 20201, USA
| | - Ziqin Zhao
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yan Jiang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Allen Burke
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; University of Maryland Medical System, Baltimore, MD 20201, USA.
| |
Collapse
|
37
|
Wang Y, Viscarra J, Kim SJ, Sul HS. Transcriptional regulation of hepatic lipogenesis. Nat Rev Mol Cell Biol 2016; 16:678-89. [PMID: 26490400 DOI: 10.1038/nrm4074] [Citation(s) in RCA: 489] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fatty acid and fat synthesis in the liver is a highly regulated metabolic pathway that is important for very low-density lipoprotein (VLDL) production and thus energy distribution to other tissues. Having common features at their promoter regions, lipogenic genes are coordinately regulated at the transcriptional level. Transcription factors, such as upstream stimulatory factors (USFs), sterol regulatory element-binding protein 1C (SREBP1C), liver X receptors (LXRs) and carbohydrate-responsive element-binding protein (ChREBP) have crucial roles in this process. Recently, insights have been gained into the signalling pathways that regulate these transcription factors. After feeding, high blood glucose and insulin levels activate lipogenic genes through several pathways, including the DNA-dependent protein kinase (DNA-PK), atypical protein kinase C (aPKC) and AKT-mTOR pathways. These pathways control the post-translational modifications of transcription factors and co-regulators, such as phosphorylation, acetylation or ubiquitylation, that affect their function, stability and/or localization. Dysregulation of lipogenesis can contribute to hepatosteatosis, which is associated with obesity and insulin resistance.
Collapse
Affiliation(s)
- Yuhui Wang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720, USA
| | - Jose Viscarra
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720, USA
| | - Sun-Joong Kim
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720, USA
| | - Hei Sook Sul
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720, USA
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Combined hyperlipidemia (CHL) is a complex phenotype that is commonly encountered clinically and is often associated with the expression of early heart disease. The affixed adjective 'familial' gives the impression that the trait is monogenic, like familial hypercholesterolemia. But despite significant efforts, genetic studies have yielded little evidence of single gene determinants of CHL. RECENT FINDINGS Sophisticated linkage studies suggest that individual lipid components of the CHL phenotype - such as elevated LDL and triglyceride - each have several determinants that segregate independently in families. Furthermore, DNA sequencing shows that rare large-effect variants in genes such as LDL receptor (LDLR) and lipoprotein lipase are found in some CHL patients, explaining the elevated LDL cholesterol and triglyceride components, respectively. In addition, multiple common small-effect lipid-altering variants accumulate in an individual's genome, raising the LDL cholesterol and/or triglyceride components by multiple mechanisms. Finally, secondary factors, such as poor diet, obesity,fatty liver or diabetes further modulate the expression of the biochemically defined CHL phenotype. SUMMARY Given the current state of genetic understanding, CHL may be best conceptualized as a syndrome with common clinical presentation but multigenic causes, similar to other common conditions such as type 2 diabetes.
Collapse
Affiliation(s)
- Amanda J Brahm
- Department of Medicine, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | | |
Collapse
|
39
|
MOCCS: Clarifying DNA-binding motif ambiguity using ChIP-Seq data. Comput Biol Chem 2016; 63:62-72. [PMID: 26971251 DOI: 10.1016/j.compbiolchem.2016.01.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 01/25/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND As a key mechanism of gene regulation, transcription factors (TFs) bind to DNA by recognizing specific short sequence patterns that are called DNA-binding motifs. A single TF can accept ambiguity within its DNA-binding motifs, which comprise both canonical (typical) and non-canonical motifs. Clarification of such DNA-binding motif ambiguity is crucial for revealing gene regulatory networks and evaluating mutations in cis-regulatory elements. Although chromatin immunoprecipitation sequencing (ChIP-seq) now provides abundant data on the genomic sequences to which a given TF binds, existing motif discovery methods are unable to directly answer whether a given TF can bind to a specific DNA-binding motif. RESULTS Here, we report a method for clarifying the DNA-binding motif ambiguity, MOCCS. Given ChIP-Seq data of any TF, MOCCS comprehensively analyzes and describes every k-mer to which that TF binds. Analysis of simulated datasets revealed that MOCCS is applicable to various ChIP-Seq datasets, requiring only a few minutes per dataset. Application to the ENCODE ChIP-Seq datasets proved that MOCCS directly evaluates whether a given TF binds to each DNA-binding motif, even if known position weight matrix models do not provide sufficient information on DNA-binding motif ambiguity. Furthermore, users are not required to provide numerous parameters or background genomic sequence models that are typically unavailable. MOCCS is implemented in Perl and R and is freely available via https://github.com/yuifu/moccs. CONCLUSIONS By complementing existing motif-discovery software, MOCCS will contribute to the basic understanding of how the genome controls diverse cellular processes via DNA-protein interactions.
Collapse
|
40
|
Yamanaka T, Tosaki A, Kurosawa M, Shimogori T, Hattori N, Nukina N. Genome-wide analyses in neuronal cells reveal that upstream transcription factors regulate lysosomal gene expression. FEBS J 2016; 283:1077-87. [DOI: 10.1111/febs.13650] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/17/2015] [Accepted: 01/08/2016] [Indexed: 12/01/2022]
Affiliation(s)
- Tomoyuki Yamanaka
- Laboratory of Structural Neuropathology; Doshisha University Graduate School of Brain Science; Kyoto Japan
- Laboratory for Structural Neuropathology; RIKEN Brain Science Institute; Saitama Japan
- Department of Neuroscience for Neurodegenerative Disorders; Juntendo University Graduate School of Medicine; Tokyo Japan
- Laboratory for Molecular Mechanisms of Thalamus Development; RIKEN Brain Science Institute; Saitama Japan
| | - Asako Tosaki
- Laboratory for Structural Neuropathology; RIKEN Brain Science Institute; Saitama Japan
| | - Masaru Kurosawa
- Laboratory for Structural Neuropathology; RIKEN Brain Science Institute; Saitama Japan
- Department of Neuroscience for Neurodegenerative Disorders; Juntendo University Graduate School of Medicine; Tokyo Japan
| | - Tomomi Shimogori
- Laboratory for Molecular Mechanisms of Thalamus Development; RIKEN Brain Science Institute; Saitama Japan
| | - Nobutaka Hattori
- Department of Neurology; Juntendo University Graduate School of Medicine; Tokyo Japan
| | - Nobuyuki Nukina
- Laboratory of Structural Neuropathology; Doshisha University Graduate School of Brain Science; Kyoto Japan
- Laboratory for Structural Neuropathology; RIKEN Brain Science Institute; Saitama Japan
- Department of Neuroscience for Neurodegenerative Disorders; Juntendo University Graduate School of Medicine; Tokyo Japan
- Laboratory for Molecular Mechanisms of Thalamus Development; RIKEN Brain Science Institute; Saitama Japan
| |
Collapse
|
41
|
Update on the molecular biology of dyslipidemias. Clin Chim Acta 2016; 454:143-85. [DOI: 10.1016/j.cca.2015.10.033] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/24/2015] [Accepted: 10/30/2015] [Indexed: 12/20/2022]
|
42
|
Ayuso M, Fernández A, Núñez Y, Benítez R, Isabel B, Barragán C, Fernández AI, Rey AI, Medrano JF, Cánovas Á, González-Bulnes A, López-Bote C, Ovilo C. Comparative Analysis of Muscle Transcriptome between Pig Genotypes Identifies Genes and Regulatory Mechanisms Associated to Growth, Fatness and Metabolism. PLoS One 2015; 10:e0145162. [PMID: 26695515 PMCID: PMC4687939 DOI: 10.1371/journal.pone.0145162] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/30/2015] [Indexed: 12/22/2022] Open
Abstract
Iberian ham production includes both purebred (IB) and Duroc-crossbred (IBxDU) Iberian pigs, which show important differences in meat quality and production traits, such as muscle growth and fatness. This experiment was conducted to investigate gene expression differences, transcriptional regulation and genetic polymorphisms that could be associated with the observed phenotypic differences between IB and IBxDU pigs. Nine IB and 10 IBxDU pigs were slaughtered at birth. Morphometric measures and blood samples were obtained and samples from Biceps femoris muscle were employed for compositional and transcriptome analysis by RNA-Seq technology. Phenotypic differences were evident at this early age, including greater body size and weight in IBxDU and greater Biceps femoris intramuscular fat and plasma cholesterol content in IB newborns. We detected 149 differentially expressed genes between IB and IBxDU neonates (p < 0.01 and Fold-Change > 1. 5). Several were related to adipose and muscle tissues development (DLK1, FGF21 or UBC). The functional interpretation of the transcriptomic differences revealed enrichment of functions and pathways related to lipid metabolism in IB and to cellular and muscle growth in IBxDU pigs. Protein catabolism, cholesterol biosynthesis and immune system were functions enriched in both genotypes. We identified transcription factors potentially affecting the observed gene expression differences. Some of them have known functions on adipogenesis (CEBPA, EGRs), lipid metabolism (PPARGC1B) and myogenesis (FOXOs, MEF2D, MYOD1), which suggest a key role in the meat quality differences existing between IB and IBxDU hams. We also identified several polymorphisms showing differential segregation between IB and IBxDU pigs. Among them, non-synonymous variants were detected in several transcription factors as PPARGC1B and TRIM63 genes, which could be associated to altered gene function. Taken together, these results provide information about candidate genes, metabolic pathways and genetic polymorphisms potentially involved in phenotypic differences between IB and IBxDU pigs associated to meat quality and production traits.
Collapse
Affiliation(s)
- Miriam Ayuso
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | | | - Yolanda Núñez
- Departamento de Mejora Genética Animal, INIA, Madrid, Spain
| | - Rita Benítez
- Departamento de Mejora Genética Animal, INIA, Madrid, Spain
| | - Beatriz Isabel
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | | | | | - Ana Isabel Rey
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | - Juan F. Medrano
- Department of Animal Science, University of California Davis, Davis, California, United States of America
| | - Ángela Cánovas
- Department of Animal Science, University of California Davis, Davis, California, United States of America
| | | | - Clemente López-Bote
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | - Cristina Ovilo
- Departamento de Mejora Genética Animal, INIA, Madrid, Spain
- * E-mail:
| |
Collapse
|
43
|
Bell DA, Watts GF. Contemporary and Novel Therapeutic Options for Hypertriglyceridemia. Clin Ther 2015; 37:2732-50. [DOI: 10.1016/j.clinthera.2015.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 07/31/2015] [Accepted: 08/05/2015] [Indexed: 12/16/2022]
|
44
|
Contribution of mutations in low density lipoprotein receptor (LDLR) and lipoprotein lipase (LPL) genes to familial combined hyperlipidemia (FCHL): A reappraisal by using a resequencing approach. Atherosclerosis 2015; 242:618-24. [DOI: 10.1016/j.atherosclerosis.2015.06.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 05/26/2015] [Accepted: 06/15/2015] [Indexed: 11/23/2022]
|
45
|
Binzer S, Stenager E, Binzer M, Kyvik KO, Hillert J, Imrell K. Genetic analysis of the isolated Faroe Islands reveals SORCS3 as a potential multiple sclerosis risk gene. Mult Scler 2015; 22:733-40. [DOI: 10.1177/1352458515602338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/21/2015] [Indexed: 11/15/2022]
Abstract
Background: In search of the missing heritability in multiple sclerosis (MS), additional approaches adding to the genetic discoveries of large genome-wide association studies are warranted. Objective: The objective of this research paper is to search for rare genetic MS risk variants in the genetically homogenous population of the isolated Faroe Islands. Methods: Twenty-nine Faroese MS cases and 28 controls were genotyped with the HumanOmniExpressExome-chip. The individuals make up 1596 pair-combinations in which we searched for identical-by-descent shared segments using the PLINK-program. Results: A segment spanning 63 SNPs with excess case-case-pair sharing was identified (0.00173 < p > 0.00212). A haplotype consisting of 42 of the 63 identified SNPs which spanned the entire the Sortilin-related vacuolar protein sorting 10 domain containing receptor 3 ( SORCS3) gene had a carrier frequency of 0.34 in cases but was not present in any controls ( p = 0.0008). Conclusion: This study revealed an oversharing in case-case-pairs of a segment spanning 63 SNPs and the entire SORCS3. While not previously associated with MS, SORCS3 appears to be important in neuronal plasticity through its binding of neurotrophin factors and involvement in glutamate homeostasis. Although additional work is needed to scrutinise the genetic effect of the SORCS3-covering haplotype, this study suggests that SORCS3 may also be important in MS pathogenesis.
Collapse
Affiliation(s)
- S Binzer
- Institute of Regional Health Research, University of Southern Denmark, Denmark/Hospital of Southern Jutland, Denmark/Odense Patient data Explorative Network (OPEN), University of Southern Denmark, Denmark/ Torshavn National Hospital, Faroe Islands
| | - E Stenager
- Institute of Regional Health Research, University of Southern Denmark, Denmark/Hospital of Southern Jutland, Denmark/ MS Clinic of Southern Jutland (Sønderborg, Esbjerg, Vejle), Department of Neurology, Denmark
| | - M Binzer
- Institute of Regional Health Research, University of Southern Denmark, Denmark
| | - KO Kyvik
- Department of Clinical Research, University of Southern Denmark, Denmark/Odense Patient data Explorative Network (OPEN), University of Southern Denmark, Denmark
| | - J Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - K Imrell
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
46
|
Niemiec P, Nowak T, Iwanicki T, Gorczynska-Kosiorz S, Balcerzyk A, Krauze J, Grzeszczak W, Wiecha M, Zak I. The rs2516839 Polymorphism of the USF1 Gene May Modulate Serum Triglyceride Levels in Response to Cigarette Smoking. Int J Mol Sci 2015; 16:13203-16. [PMID: 26068452 PMCID: PMC4490492 DOI: 10.3390/ijms160613203] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 05/30/2015] [Accepted: 06/01/2015] [Indexed: 01/29/2023] Open
Abstract
Single nucleotide polymorphisms (SNPs) of the USF1 gene (upstream stimulatory factor 1) influence plasma lipid levels. This study aims to determine whether USF1 SNPs interact with traditional risk factors of atherosclerosis to increase coronary artery disease (CAD) risk. In the present study serum lipid levels and USF1 gene polymorphisms (rs2516839 and rs3737787) were determined in 470 subjects: 235 patients with premature CAD and 235 controls. A trend of increasing triglycerides (TG) levels in relation to the C allele dose of rs2516839 SNP was observed. The synergistic effect of cigarette smoking and C allele carrier state on CAD risk was also found (SIM = 2.69, p = 0.015). TG levels differentiated significantly particular genotypes in smokers (1.53 mmol/L for TT, 1.80 mmol/L for CT and 2.27 mmol/L for CC subjects). In contrast, these differences were not observed in the non-smokers subgroup (1.57 mmol/L for TT, 1.46 mmol/L for CT and 1.49 mmol/L for CC subjects). In conclusion, the rs2516839 polymorphism may modulate serum triglyceride levels in response to cigarette smoking. Carriers of the C allele seem to be particularly at risk of CAD, when exposed to cigarette smoking.
Collapse
Affiliation(s)
- Pawel Niemiec
- School of Health Sciences in Katowice, Medical University of Silesia, Department of Biochemistry and Medical Genetics, Medykow Str 18, 40-752 Katowice, Poland.
| | - Tomasz Nowak
- School of Health Sciences in Katowice, Medical University of Silesia, Department of Biochemistry and Medical Genetics, Medykow Str 18, 40-752 Katowice, Poland.
| | - Tomasz Iwanicki
- School of Health Sciences in Katowice, Medical University of Silesia, Department of Biochemistry and Medical Genetics, Medykow Str 18, 40-752 Katowice, Poland.
| | - Sylwia Gorczynska-Kosiorz
- School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Department of Internal Medicine, Diabetes and Nephrology, 3 Maja Str 13-18, 41-800 Zabrze, Poland.
| | - Anna Balcerzyk
- School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Department of Internal Medicine, Diabetes and Nephrology, 3 Maja Str 13-18, 41-800 Zabrze, Poland.
| | - Jolanta Krauze
- School of Medicine in Katowice, Medical University of Silesia, 1st Department of Cardiac Surgery in Upper Silesian Center of Cardiology in Katowice, Ziolowa Str 47, 40-635 Katowice, Poland.
| | - Wladyslaw Grzeszczak
- School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Department of Internal Medicine, Diabetes and Nephrology, 3 Maja Str 13-18, 41-800 Zabrze, Poland.
| | - Maria Wiecha
- Regional Centre of Blood Donation and Blood Treatment in Raciborz, Sienkiewicza Str 3, 47-400 Raciborz, Poland.
| | - Iwona Zak
- School of Health Sciences in Katowice, Medical University of Silesia, Department of Biochemistry and Medical Genetics, Medykow Str 18, 40-752 Katowice, Poland.
| |
Collapse
|
47
|
Whayne TF, Parinandi N, Maulik N. Thioredoxins in cardiovascular disease. Can J Physiol Pharmacol 2015; 93:903-11. [PMID: 26417924 DOI: 10.1139/cjpp-2015-0105] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Key thioredoxin (Trx) system components are nicotinamide adenine dinucleotide phosphate (NADPH), Trx reductase (TrxR), and Trx. TrxR catalyzes disulfide reduction in Trx with NADPH as cofactor. Because Trx is an antioxidant, oxidative stress results in an increase in Trx, which has a reduced disulfide component. If Trx is suppressed, oxidative stress in higher. In contrast a decrease in oxidative stress is associated with low Trx levels. Trx is involved in inflammation, apoptosis, embryogenesis, and cardiovascular disease (CVD). This review focuses on the Trx system in CVD. Abnormal Trx binding occurs in mouse familial combined hyperlipidemia; however, this has not been confirmed in humans. Congestive heart failure is a manifestation of many CVDs, which may be improved by attenuating oxidative stress through the suppression of Trx and decreased reactive oxygen species. Angiotensin II is associated with hypertension and other CVDs, and its receptor blockade results in decreased oxidative stress with reduced Trx levels. Inflammation is a major causative factor of CVDs, and myocarditis as an example, is associated with increased Trx levels. Vascular endothelial dysfunction has an association with CVD. This dysfunction is alleviated by hormone replacement therapy, which involves decreased oxidative stress and Trx levels. Diabetes mellitus has a major association with CVDs; increase in Trx levels may reflect insulin resistance. Identification of Trx system abnormalities may lead to innovative approaches to treat multiple CVDs and other pathologies.
Collapse
Affiliation(s)
- Thomas F Whayne
- a Gill Heart Institute, University of Kentucky, 326 Wethington Building, 900 South Limestone Street, Lexington, KY 40536-0200, USA
| | - Narasimham Parinandi
- b Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Nilanjana Maulik
- c Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
48
|
Hernando Boigues JF, Mach N. The effect of polyunsaturated fatty acids on obesity through epigenetic modifications. ACTA ACUST UNITED AC 2015; 62:338-49. [PMID: 26003266 DOI: 10.1016/j.endonu.2015.03.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 03/17/2015] [Accepted: 03/17/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE In recent years it has been demonstrated that polyunsaturated fatty acids (PUFA) have anti-inflammatory and as regulators of lipid metabolism. However, the epigenomic mechanisms involved in these processes are not known in depth. The aim of this review was to describe the scientific evidence supports that regular consumption of PUFA may reduce obesity and overweight by altering epigenetic marks. MATERIAL AND METHODS A search of recent publications was carried out in human clinical trials, as well as animal model and in vitro experiments. RESULTS Exist a possible therapeutic effect of PUFAs on the prevention and development of obesity due to their ability to reversively modify the methylation of the promoters of genes associated with lipid metabolism and to modulate the activity of certain microRNAs. CONCLUSIONS A better knowledge and understanding of the PUFAs role in epigenetic regulation of obesity is possible with the current published results. The PUFAs may modulate the promotor epigenetic marks in several adipogenic genes and regulate the expression of several miRNAs.
Collapse
Affiliation(s)
- Julián F Hernando Boigues
- Àrea de Ciències de la Salut, Institut Internacional de Postgrau, Universitat Oberta de Catalunya (UOC), Barcelona, España
| | - Núria Mach
- Àrea de Ciències de la Salut, Institut Internacional de Postgrau, Universitat Oberta de Catalunya (UOC), Barcelona, España; INRA, Animal Genetics and Integrative Biology Unit, Jouy-en-Josas, Francia.
| |
Collapse
|
49
|
Blackett PR, Wilson DP, McNeal CJ. Secondary hypertriglyceridemia in children and adolescents. J Clin Lipidol 2015; 9:S29-40. [PMID: 26343210 DOI: 10.1016/j.jacl.2015.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/22/2015] [Accepted: 04/22/2015] [Indexed: 12/22/2022]
Abstract
Secondary dyslipidemia with predominant hypertriglyceridemia may occur as a consequence of both common and rare causes. After accounting for obesity and associated insulin resistance, clinicians should carefully consider other contributing factors and conditions. Genetic background and causative factors prevail during gestation, infancy, and childhood and continue in adults. Elevations in triglyceride (TG) are associated with transfer of TG to high-density lipoprotein (HDL) and low-density lipoprotein (LDL) resulting in lipolysis, HDL degradation, and formation of atherogenic LDL particles. Defining and treating the underlying cause is the first step toward restoring the lipids and lipoproteins to normal, especially in cases with severe hypertriglyceridemia, who are at risk for acute pancreatitis. Disorders involving the liver, kidney, endocrine, and immune systems and medications need to be considered. Rare diseases such as lipodystrophy and glycogen storage disease are particularly challenging, and there have been promising new developments. Treatment depends on the severity; prevention of acute pancreatitis being a priority in severe cases and lifestyle modification being a foundation for general management followed by targeting TG and predictors of coronary artery disease such as LDL cholesterol and non-HDL cholesterol, when they exceed cutpoints.
Collapse
Affiliation(s)
- Piers R Blackett
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Don P Wilson
- Department of Pediatrics, Pediatric Endocrinology and Diabetes, Cook Children's Medical Center, Fort Worth, TX, USA
| | - Catherine J McNeal
- Department of Pediatrics, Scott & White Healthcare, Temple, TX, USA; Division of Cardiology, Department of Internal Medicine, Scott & White Healthcare, Temple, TX, USA
| |
Collapse
|
50
|
Luo X, Yu C, Fu C, Shi W, Wang X, Zeng C, Wang H. Identification of the differentially expressed genes associated with familial combined hyperlipidemia using bioinformatics analysis. Mol Med Rep 2015; 11:4032-8. [PMID: 25625967 PMCID: PMC4394960 DOI: 10.3892/mmr.2015.3263] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 11/18/2014] [Indexed: 12/21/2022] Open
Abstract
The aim of the present study was to screen the differentially expressed genes (DEGs) associated with familial combined hyperlipidemia (FCHL) and examine the changing patterns. The transcription profile of GSE18965 was obtained from the NCBI Gene Expression Omnibus database, including 12 FCHL samples and 12 control specimens. The DEGs were identified using a linear models for microarray data package in the R programming language. Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was also performed. Protein-protein interaction (PPI) networks of the DEGs were constructed using the EnrichNet online tool. In addition, cluster analysis of the genes in networks was performed using ClusterONE. A total of 879 DEGs were screened, including 394 upregulated and 485 downregulated genes. Enrichment analysis identified four important KEGG pathways associated with FCHL: One carbon pool by folate, α-linolenic acid metabolism, asthma and the glycosphingolipid biosynthesis-globo series. GO annotation identified 12 enriched biological processes, including one associated with hematopoiesis and four associated with bone cell differentiation. This identification was in accordance with clinical data and experiments into hyperlipidemia and bone lesions. Based on PPI networks, these DEGs had a close association with immune responses, hormone responses and cytokine-cytokine receptors. In conclusion, these DEGs may be used as specific therapeutic molecular targets in the treatment of FCHL. The present findings may provide the basis for understanding the pathogenesis of FCHL in future studies. However, further experiments are required to confirm these results.
Collapse
Affiliation(s)
- Xiaoli Luo
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Changqing Yu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Chunjiang Fu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Weibin Shi
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Xukai Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Hongyong Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| |
Collapse
|