1
|
Sakurai-Yageta M, Suzuki Y. Molecular Mechanisms of Biotin in Modulating Inflammatory Diseases. Nutrients 2024; 16:2444. [PMID: 39125325 PMCID: PMC11314543 DOI: 10.3390/nu16152444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Biotin, also known as vitamin B7 or vitamin H, is a water-soluble B-complex vitamin and serves as an essential co-enzyme for five specific carboxylases. Holocarboxylase synthase (HCS) activates biotin and facilitates its covalent attachment to these enzymes, while biotinidase releases free biotin in the biotin cycle. The transport of biotin, primarily from the intestine, is mediated by the sodium-dependent multi-vitamin transporter (SMVT). Severe biotin deficiency leads to multiple carboxylase deficiency. Moreover, biotin is crucial to glucose and lipid utilization in cellular energy production because it modulates the expression of metabolic enzymes via various signaling pathways and transcription factors. Biotin also modulates the production of proinflammatory cytokines in the immune system through similar molecular mechanisms. These regulatory roles in metabolic and immune homeostasis connect biotin to conditions such as diabetes, dermatologic manifestations, and multiple sclerosis. Furthermore, deficiencies in biotin and SMVT are implicated in inflammatory bowel disease, affecting intestinal inflammation, permeability, and flora. Notably, HCS and probably biotin directly influence gene expression through histone modification. In this review, we summarize the current knowledge on the molecular aspects of biotin and associated molecules in diseases related to both acute inflammatory responses and chronic inflammation, and discuss the potential therapeutic applications of biotin.
Collapse
Affiliation(s)
- Mika Sakurai-Yageta
- Department of Education and Training, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Miyagi, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai 980-8573, Miyagi, Japan
| | - Yoichi Suzuki
- Department of Education and Training, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Miyagi, Japan
- Department of Clinical Genetics, Ageo Central General Hospital, Ageo 362-8588, Saitama, Japan
| |
Collapse
|
2
|
Yan L, Li J, Hu J, Qu J, Li K, Wang M, An SS, Ke CC, Li H, Yuan F, Guo W, Hu M, Zhang J, Yang Z, Mu H, zhang F, Zhang J, Cui X, Hu Y. Biotin attenuates heat shock factor 4b transcriptional activity by lysine 444 biotinylation. Biochem Biophys Rep 2022; 30:101227. [PMID: 35198740 PMCID: PMC8841385 DOI: 10.1016/j.bbrep.2022.101227] [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: 11/22/2021] [Revised: 01/16/2022] [Accepted: 01/31/2022] [Indexed: 11/16/2022] Open
Abstract
Genetic mutations in HSF4 cause congenital cataracts. HSF4 exhibits both positive and negative regulation on the transcription of heat shock and non-heat shock proteins during lens development, and its activity is regulated by posttranslational modifications. Biotin is an essential vitamin that regulates gene expression through protein biotinylation. In this paper, we report that HSF4b is negatively regulated by biotinylation. Administration of biotin or ectopic bacterial biotin ligase BirA increases HSF4b biotinylation at its C-terminal amino acids from 196 to 493. This attenuates the HSF4b-controlled expression of αB-crystallin in both lens epithelial cells and tested HEK293T cells. HSF4b interacts with holocarboxylase synthetase (HCS), a ubiquitous enzyme for catalyzing protein biotinylation in mammal. Ectopic HA-HCS expression downregulates HSF4b-controlled αB-crystallin expression. Lysine-mutation analyses indicate that HSF4b/K444 is a potential biotinylation site. Mutation K444R reduces the co-precipitation of HSF4b by streptavidin beads and biotin-induced reduction of αB-crystallin expression. Mutations of other lysine residues such as K207R/K209R, K225R, K288R, K294R and K355R in HSF4's C-terminal region do not affect HSF4's expression level and the interaction with streptavidin, but they exhibit distinct regulation on αB-crystallin expression through different mechanisms. HSF4/K294R leads to upregulation of αB-crystallin expression, while mutations K207R/K209R, K225R, K288R, K255R and K435R attenuate HSF4's regulation on αB-crystallin expression. K207R/K209R blocks HSF4 nuclear translocation, and K345R causes HSF4 destabilization. Taken together, the data reveal that biotin maybe a novel factor in modulating HSF4 activity through biotinylation. Biotin downregulates HSF4's transcription activity. HSF4 is associated with and down-regulated by holocarboxylase synthetase (HCS). K444 is the potential biotinylated amino acid residue in HSF4b.
Collapse
Affiliation(s)
- Longjun Yan
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Jing Li
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Jialin Hu
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Junwei Qu
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Kejia Li
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Mingli Wang
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Shuang-Shuang An
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Cun-cun Ke
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Hui Li
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Fengling Yuan
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Weikai Guo
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Mengyue Hu
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Jing Zhang
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Zhengyan Yang
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Hongmei Mu
- Kaifeng Key Lab for Cataract and Myopia, Institute of Eye Disease, Kaifeng Central Hospital, Kaifeng, China
| | - Fengyan zhang
- Department of Ophthalmology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun Zhang
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
| | - Xiukun Cui
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
- Corresponding author.
| | - Yanzhong Hu
- National-Joint Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan International Union Lab of Antibody Medicine, Department of Cell Biology and Genetics, Henan University School of Basic Medical Sciences, Kaifeng, China
- Department of Ophthalmology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Kaifeng Key Lab for Cataract and Myopia, Institute of Eye Disease, Kaifeng Central Hospital, Kaifeng, China
- Corresponding author. Department of Cell Biology, Henan University School of Medicine, Zhengzhou, China.
| |
Collapse
|
3
|
Biotin rescues mitochondrial dysfunction and neurotoxicity in a tauopathy model. Proc Natl Acad Sci U S A 2020; 117:33608-33618. [PMID: 33318181 DOI: 10.1073/pnas.1922392117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mitochondrial and metabolic dysfunction are often implicated in neurological disease, but effective mechanism-based therapies remain elusive. We performed a genome-scale forward genetic screen in a Drosophila model of tauopathy, a class of neurodegenerative disorders characterized by the accumulation of the protein tau, and identified manipulation of the B-vitamin biotin as a potential therapeutic approach in tauopathy. We show that tau transgenic flies have an innate biotin deficiency due to tau-mediated relaxation of chromatin and consequent aberrant expression of multiple biotin-related genes, disrupting both carboxylase and mitochondrial function. Biotin depletion alone causes mitochondrial pathology and neurodegeneration in both flies and human neurons, implicating mitochondrial dysfunction as a mechanism in biotin deficiency. Finally, carboxylase biotin levels are reduced in mammalian tauopathies, including brains of human Alzheimer's disease patients. These results provide insight into pathogenic mechanisms of human biotin deficiency, the resulting effects on neuronal health, and a potential therapeutic pathway in the treatment of tau-mediated neurotoxicity.
Collapse
|
4
|
Watts JS, Morton DG, Kemphues KJ, Watts JL. The biotin-ligating protein BPL-1 is critical for lipid biosynthesis and polarization of the Caenorhabditis elegans embryo. J Biol Chem 2018; 293:610-622. [PMID: 29158261 PMCID: PMC5767866 DOI: 10.1074/jbc.m117.798553] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 11/16/2017] [Indexed: 01/07/2023] Open
Abstract
Biotin is an essential cofactor for multiple metabolic reactions catalyzed by carboxylases. Biotin is covalently linked to apoproteins by holocarboxylase synthetase (HCS). Accordingly, some mutations in HCS cause holocarboxylase deficiency, a rare metabolic disorder that can be life-threatening if left untreated. However, the long-term effects of HCS deficiency are poorly understood. Here, we report our investigations of bpl-1, which encodes the Caenorhabditis elegans ortholog of HCS. We found that mutations in the biotin-binding region of bpl-1 are maternal-effect lethal and cause defects in embryonic polarity establishment, meiosis, and the integrity of the eggshell permeability barrier. We confirmed that BPL-1 biotinylates four carboxylase enzymes, and we demonstrate that BPL-1 is required for efficient de novo fatty acid biosynthesis. We also show that the lack of larval growth defects as well as nearly normal fatty acid composition in young adult worms is due to sufficient fatty acid precursors provided by dietary bacteria. However, BPL-1 disruption strongly decreased levels of polyunsaturated fatty acids in embryos produced by bpl-1 mutant hermaphrodites, revealing a critical role for BPL-1 in lipid biosynthesis during embryogenesis and demonstrating that dietary fatty acids and lipid precursors are not adequate to support early embryogenesis in the absence of BPL-1. Our findings highlight that studying BPL-1 function in C. elegans could help dissect the roles of this important metabolic enzyme under different environmental and dietary conditions.
Collapse
Affiliation(s)
- Jason S Watts
- From the School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, Washington 99164-7520 and
| | - Diane G Morton
- the Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14850
| | - Kenneth J Kemphues
- the Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14850
| | - Jennifer L Watts
- From the School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, Washington 99164-7520 and
| |
Collapse
|
5
|
Choi HW, Shin PG, Lee JH, Choi WS, Kang MJ, Kong WS, Oh MJ, Seo YB, Kim GD. Anti-inflammatory effect of lovastatin is mediated via the modulation of NF-κB and inhibition of HDAC1 and the PI3K/Akt/mTOR pathway in RAW264.7 macrophages. Int J Mol Med 2017; 41:1103-1109. [PMID: 29207042 DOI: 10.3892/ijmm.2017.3309] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 11/15/2017] [Indexed: 11/06/2022] Open
Abstract
Lovastatin is a 3-hydroxy-3-methylglutaryl-CoA reductase inhibitor that is clinically used for the prevention of cardiovascular diseases. Although it has been reported that lovastatin has anti-inflammatory properties in several studies, how lovastatin regulates the inflammation is still unclear. To evaluate the effect of lovastatin on nitric oxide production (NO) in RAW264.7 macrophages, NO production assay was performed. Also, cell viability was measured to confirm cytotoxicity. Level of tumor necrosis factor-α (TNF-α) transcription was measured by reverse transcription polymerase chain reaction (RT-PCR) from total RNA in RAW264.7 cells. Western blot analysis and immunofluorescence staining were used to investigate the regulation of lovastatin on the expression, phosphorylation, and nuclear translocation of cellular proteins. The results of the present study revealed that lovastatin reduced nitric oxide production via the reduction of inducible nitric oxide synthase (iNOS) expression in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophage cells. The mRNA level of TNF-α was reduced in presence of lovastatin. In addition, lovastatin downregulated histone deacetylase 1 (HDAC1), resulting in the accumulation of acetylated histone H3 and heat shock protein 70. Furthermore, the expression of phosphoinositide 3-kinase catalytic subunits α and β was reduced under lovastatin treatment, and the phosphorylation of Akt and mammalian target of rapamycin was consequently inhibited. Lovastatin also inhibited the phosphorylation of inhibitor of nuclear factor (NF)-κBα and the translocation of NF-κB into the nucleus. Therefore, the present study demonstrates that lovastatin inhibits the expression of pro-inflammatory mediators, including iNOS and TNF-α, through the suppression of HDAC1 expression, PI3K/Akt phosphorylation and NF-κB translocation in LPS-stimulated RAW264.7 macrophage cells.
Collapse
Affiliation(s)
- Hyung-Wook Choi
- Department of Microbiology, Pukyong National University, Busan 48513, Republic of Korea
| | - Pyung-Gyun Shin
- Mushroom Research Division, National Institute of Horticultural and Herbal Science, Rural Development Administration, Eumseong 27709, Republic of Korea
| | - Ji-Hyun Lee
- Department of Microbiology, Pukyong National University, Busan 48513, Republic of Korea
| | - Woo-Suk Choi
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Min-Jae Kang
- Department of Microbiology, Pukyong National University, Busan 48513, Republic of Korea
| | - Won-Sik Kong
- Mushroom Research Division, National Institute of Horticultural and Herbal Science, Rural Development Administration, Eumseong 27709, Republic of Korea
| | - Min-Ji Oh
- Mushroom Research Division, National Institute of Horticultural and Herbal Science, Rural Development Administration, Eumseong 27709, Republic of Korea
| | - Yong-Bae Seo
- Department of Microbiology, Pukyong National University, Busan 48513, Republic of Korea
| | - Gun-Do Kim
- Department of Microbiology, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
6
|
León-Del-Río A, Valadez-Graham V, Gravel RA. Holocarboxylase Synthetase: A Moonlighting Transcriptional Coregulator of Gene Expression and a Cytosolic Regulator of Biotin Utilization. Annu Rev Nutr 2017; 37:207-223. [DOI: 10.1146/annurev-nutr-042617-104653] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Alfonso León-Del-Río
- Programa de Investigación de Cáncer de Mama y Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de Mexico 04500, México
| | - Viviana Valadez-Graham
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62250, México
| | - Roy A. Gravel
- Department of Biochemistry & Molecular Biology, the University of Calgary and the Alberta Children's Hospital Research Institute for Child and Maternal Health, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
7
|
Xia B, Yang LQ, Huang HY, Pang L, Yang XF, Yi YJ, Ren XH, Li J, Zhuang ZX, Liu JJ. Repression of Biotin-Related Proteins by Benzo[a]Pyrene-Induced Epigenetic Modifications in Human Bronchial Epithelial Cells. Int J Toxicol 2016; 35:336-43. [PMID: 26960346 DOI: 10.1177/1091581816637071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Benzo[a]pyrene (B[a]P) exposure has been associated with the alteration in epigenetic marks that are involved in cancer development. Biotinidase (BTD) and holocarboxylase synthetase (HCS) are 2 major enzymes involved in maintaining the homeostasis of biotinylation, and the deregulation of this pathway has been associated with a number of cancers. However, the link between B[a]P exposure and the dysregulation of BTD/HCS in B[a]P-associated tumorigenesis is unknown. Here we showed that the expression of both BTD and HCS was significantly decreased upon B[a]P treatment in human bronchial epithelial (16HBE) cells. Benzo[a]pyrene exposure led to the global loss of DNA methylation by immunofluorescence, which coincided with the reduction in acetylation levels on histones H3 and H4 in 16HBE cells. Consistent with decreased histone acetylation, histone deacetylases (HDACs) HDAC2 and HDAC3 were significantly upregulated in a dosage-dependent manner. When DNA methylation or HDAC activity was inhibited, we found that the reduction in BTD and HCS was separately regulated through distinct epigenetic mechanisms. Together, our results suggested the potential link between B[a]P toxicity and deregulation of biotin homeostasis pathway in B[a]P-associated cancer development.
Collapse
Affiliation(s)
- Bo Xia
- Key Laboratory of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China College of Food Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Lin-Qing Yang
- Key Laboratory of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Hai-Yan Huang
- Key Laboratory of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Li Pang
- College of Horticulture and Gardening, Hunan Agricultural University, Changsha, Hunan, China
| | - Xi-Fei Yang
- Key Laboratory of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - You-Jin Yi
- College of Food Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
| | - Xiao-Hu Ren
- Key Laboratory of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Jie Li
- Key Laboratory of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Zhi-Xiong Zhuang
- Key Laboratory of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Jian-Jun Liu
- Key Laboratory of Modern Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| |
Collapse
|
8
|
Biotin-mediated epigenetic modifications: Potential defense against the carcinogenicity of benzo[a]pyrene. Toxicol Lett 2016; 241:216-24. [DOI: 10.1016/j.toxlet.2015.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/03/2015] [Accepted: 11/08/2015] [Indexed: 12/16/2022]
|
9
|
Malashicheva A, Bogdanova M, Zabirnyk A, Smolina N, Ignatieva E, Freilikhman O, Fedorov A, Dmitrieva R, Sjöberg G, Sejersen T, Kostareva A. Various lamin A/C mutations alter expression profile of mesenchymal stem cells in mutation specific manner. Mol Genet Metab 2015; 115:118-27. [PMID: 25982065 DOI: 10.1016/j.ymgme.2015.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 04/27/2015] [Accepted: 04/28/2015] [Indexed: 12/31/2022]
Abstract
Various mutations in LMNA gene, encoding for nuclear lamin A/C protein, lead to laminopathies and contribute to over ten human disorders, mostly affecting tissues of mesenchymal origin such as fat tissue, muscle tissue, and bones. Recently it was demonstrated that lamins not only play a structural role providing communication between extra-nuclear structures and components of cell nucleus but also control cell fate and differentiation. In our study we assessed the effect of various LMNA mutations on the expression profile of mesenchymal multipotent stem cells (MMSC) during adipogenic and osteogenic differentiation. We used lentiviral approach to modify human MMSC with LMNA-constructs bearing mutations associated with different laminopathies--G465D, R482L, G232E, R527C, and R471C. The impact of various mutations on MMSC differentiation properties and expression profile was assessed by colony-forming unit analysis, histological staining, expression of the key differentiation markers promoting adipogenesis and osteogenesis followed by the analysis of the whole set of genes involved in lineage-specific differentiation using PCR expression arrays. We demonstrate that various LMNA mutations influence the differentiation efficacy of MMSC in mutation-specific manner. Each LMNA mutation promotes a unique expression pattern of genes involved in a lineage-specific differentiation and this pattern is shared by the phenotype-specific mutations.
Collapse
Affiliation(s)
- Anna Malashicheva
- Almazov Federal Medical Research Centre, St. Petersburg, Russia; St. Petersburg State University, St. Petersburg, Russia; ITMO University, Institute of translational Medicine, St. Petersburg, Russia
| | - Maria Bogdanova
- Almazov Federal Medical Research Centre, St. Petersburg, Russia; St. Petersburg State University, St. Petersburg, Russia
| | | | - Natalia Smolina
- Almazov Federal Medical Research Centre, St. Petersburg, Russia; Department of Woman and Child Health, Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Elena Ignatieva
- Almazov Federal Medical Research Centre, St. Petersburg, Russia
| | | | - Anton Fedorov
- Almazov Federal Medical Research Centre, St. Petersburg, Russia
| | | | - Gunnar Sjöberg
- Department of Woman and Child Health, Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Thomas Sejersen
- Department of Woman and Child Health, Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Anna Kostareva
- Almazov Federal Medical Research Centre, St. Petersburg, Russia; Department of Woman and Child Health, Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden; ITMO University, Institute of translational Medicine, St. Petersburg, Russia.
| |
Collapse
|
10
|
Gravel RA. Holocarboxylase synthetase: a multitalented protein with roles in biotin transfer, gene regulation and chromatin dynamics. Mol Genet Metab 2014; 111:305-306. [PMID: 24361214 DOI: 10.1016/j.ymgme.2013.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 11/16/2013] [Indexed: 11/27/2022]
Affiliation(s)
- Roy A Gravel
- Departments of Biochemistry & Molecular Biology and of Medical Genetics and the Alberta Children's Hospital Research Institute for Child and Maternal Health, Faculties of Medicine and Kinesiology, University of Calgary, Room 250 Heritage Medical Research Building, 3330 Hospital Drive N.W., Calgary, Alberta, Canada T3H-1C1.
| |
Collapse
|