1
|
Cheng H, Zhang X, Li Y, Cao D, Luo C, Zhang Q, Zhang S, Jiao Y. Age-related testosterone decline: mechanisms and intervention strategies. Reprod Biol Endocrinol 2024; 22:144. [PMID: 39543598 PMCID: PMC11562514 DOI: 10.1186/s12958-024-01316-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Contemporary societies exhibit delayed reproductive age and increased life expectancy. While the male reproductive system demonstrates relatively delayed aging compared to that of females, increasing age substantially impacts its function. A characteristic manifestation is age-induced testosterone decline. Testosterone, a crucial male sex hormone, plays pivotal roles in spermatogenesis and sexual function, and contributes significantly to metabolism, psychology, and cardiovascular health. Aging exerts profound effects on the hypothalamic-pituitary-gonadal axis and Leydig cells, precipitating testosterone reduction, which adversely affects male health. Exogenous testosterone supplementation can partially ameliorate age-related testosterone deficiency; however, its long-term safety remains contentious. Preserving endogenous testosterone production capacity during the aging process warrants further investigation as a potential intervention strategy.
Collapse
Affiliation(s)
- Haoyang Cheng
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoyan Zhang
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi, China
| | - Yongheng Li
- Jiading Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Dezhong Cao
- First People's Hospital of Dongcheng District, Beijing, China
| | - Chenglong Luo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Sizheng Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yongzheng Jiao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
2
|
Yan S, Wang Y, Wang B, Zuo S, Yu Y. Thromboxane A 2 Modulates de novo Synthesis of Adrenal Corticosterone in Mice via p38/14-3-3γ/StAR Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307926. [PMID: 38460156 PMCID: PMC11095200 DOI: 10.1002/advs.202307926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/21/2024] [Indexed: 03/11/2024]
Abstract
Prostanoids are endogenous lipid bioactive mediators that play essential roles in physiological processes such as glucocorticoid secretion. Here, it is found that the thromboxane (Tx)A2 receptor (TP) is highly expressed in the adrenal cortex of mice. Both global and adrenocortical-specific deletion of the TP receptor lead to increased adiposity in mice by elevating corticosterone synthesis. Mechanistically, the TP receptor deletion increases the phosphorylation of steroidogenic acute regulatory protein (StAR) and corticosterone synthesis in adrenal cortical cells by suppressing p-p38-mediated phosphorylation of 14-3-3γ adapter protein at S71. The activation of the p38 in the adrenal cortical cells by forced expression of the MKK6EE gene attenuates hypercortisolism in TP-deficient mice. These observations suggest that the TxA2/TP signaling regulates adrenal corticosterone homeostasis independent of the hypothalamic-pituitary-adrenal axis and the TP receptor may serve as a promising therapeutic target for hypercortisolism.
Collapse
Affiliation(s)
- Shuai Yan
- Department of PharmacologyTianjin Key Laboratory of Inflammatory BiologyState Key Laboratory of Experimental HematologyKey Laboratory of Immune Microenvironment and Disease (Ministry of Education)The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical SciencesTianjin Medical UniversityTianjin300070P. R. China
- Division of Endocrinology, Diabetes, and MetabolismBeth Israel Deaconess Medical CenterHarvard Medical School330 Brookline AvenueBostonMassachusetts02115USA
| | - Yuanyang Wang
- Department of PharmacologyTianjin Key Laboratory of Inflammatory BiologyState Key Laboratory of Experimental HematologyKey Laboratory of Immune Microenvironment and Disease (Ministry of Education)The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical SciencesTianjin Medical UniversityTianjin300070P. R. China
| | - Bei Wang
- Department of PharmacologyTianjin Key Laboratory of Inflammatory BiologyState Key Laboratory of Experimental HematologyKey Laboratory of Immune Microenvironment and Disease (Ministry of Education)The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical SciencesTianjin Medical UniversityTianjin300070P. R. China
| | - Shengkai Zuo
- Department of PharmacologyTianjin Key Laboratory of Inflammatory BiologyState Key Laboratory of Experimental HematologyKey Laboratory of Immune Microenvironment and Disease (Ministry of Education)The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical SciencesTianjin Medical UniversityTianjin300070P. R. China
- Department of BiopharmaceuticsTianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070P. R. China
| | - Ying Yu
- Department of PharmacologyTianjin Key Laboratory of Inflammatory BiologyState Key Laboratory of Experimental HematologyKey Laboratory of Immune Microenvironment and Disease (Ministry of Education)The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsSchool of Basic Medical SciencesTianjin Medical UniversityTianjin300070P. R. China
| |
Collapse
|
3
|
Qian P, Ma F, Zhang W, Cao D, Li L, Liu Z, Pei P, Zhang T, Wang S, Wu J. Chronic exercise remodels the lysine acetylome in the mouse hippocampus. Front Mol Neurosci 2022; 15:1023482. [PMID: 36385767 PMCID: PMC9650339 DOI: 10.3389/fnmol.2022.1023482] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/10/2022] [Indexed: 11/28/2022] Open
Abstract
Physical exercise benefits hippocampal function through various molecular mechanisms. Protein acetylation, a conserved and widespread post-translational modification, is involved in the synaptic plasticity and memory. However, whether exercise can change global acetylation and the role of acetylated proteins in the hippocampus have remained largely unknown. Herein, using healthy adult mice running for 6 weeks as exercise model and sedentary mice as control, we analyzed the hippocampal lysine acetylome and proteome by Liquid chromatography-tandem mass spectrometry. As a result, we profiled the lysine acetylation landscape for the hippocampus and identified 3,876 acetyl sites and 1,764 acetylated proteins. A total of 272 acetyl sites on 252 proteins were differentially regulated by chronic exercise, among which 18.58% acetylated proteins were annotated in mitochondria. These proteins were dominantly deacetylated and mainly associated with carbon-related metabolism, the Hippo signaling pathway, ribosomes, and protein processing. Meanwhile, 21 proteins were significantly expressed and enriched in the pathway of complement and coagulation cascades. Our findings provide a new avenue for understanding the molecular mechanisms underlying the benefits of exercise for hippocampal function and can contribute to the promotion of public health.
Collapse
Affiliation(s)
- Ping Qian
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Feifei Ma
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Wanyu Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Dingding Cao
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Luya Li
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Zhuo Liu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Pei Pei
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Ting Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Shan Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
- *Correspondence: Shan Wang,
| | - Jianxin Wu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Jianxin Wu,
| |
Collapse
|
4
|
Sluchanko NN. Recent advances in structural studies of 14-3-3 protein complexes. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 130:289-324. [PMID: 35534110 DOI: 10.1016/bs.apcsb.2021.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Being phosphopeptide-binding hubs, 14-3-3 proteins coordinate multiple cellular processes in eukaryotes, including the regulation of apoptosis, cell cycle, ion channels trafficking, transcription, signal transduction, and hormone biosynthesis. Forming constitutive α-helical dimers, 14-3-3 proteins predominantly recognize specifically phosphorylated Ser/Thr sites within their partners; this generally stabilizes phosphotarget conformation and affects its activity, intracellular distribution, dephosphorylation, degradation and interactions with other proteins. Not surprisingly, 14-3-3 complexes are involved in the development of a range of diseases and are considered promising drug targets. The wide interactome of 14-3-3 proteins encompasses hundreds of different phosphoproteins, for many of which the interaction is well-documented in vitro and in vivo but lack the structural data that would help better understand underlying regulatory mechanisms and develop new drugs. Despite obtaining structural information on 14-3-3 complexes is still lagging behind the research of 14-3-3 interactions on a proteome-wide scale, recent works provided some advances, including methodological improvements and accumulation of new interesting structural data, that are discussed in this review.
Collapse
Affiliation(s)
- Nikolai N Sluchanko
- A.N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology" of the Russian Academy of Sciences, Moscow, Russian Federation.
| |
Collapse
|
5
|
Frontini-López YR, Gojanovich AD, Del Veliz S, Uhart M, Bustos DM. 14-3-3β isoform is specifically acetylated at Lys51 during differentiation to the osteogenic lineage. J Cell Biochem 2021; 122:1767-1780. [PMID: 34379822 DOI: 10.1002/jcb.30128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/12/2021] [Accepted: 08/03/2021] [Indexed: 01/01/2023]
Abstract
The 14-3-3 protein family binds and regulates hundreds of serine/threonine phosphorylated proteins as an essential component of many signaling networks. Specific biological functions are currently been discovered for each of its seven isoforms in mammals. These proteins have been traditionally considered unregulated; however, its acetylation in an essential lysine residue, causing its inactivation, was recently published. Here, we studied the acetylation state of this lysine 49/51 during the osteogenic differentiation of human adipose-derived stem cells. We found that during this process, the levels of 14-3-3β (but not its isoform 14-3-3γ) acK49/51 increase, representing the first report linking this PTM to a specific isoform and a cellular process. Our results suggested that this posttranslational modification could be catalyzed by the HBO1 acetyltransferase, as overexpression of HBO1 increased specifically 14-3-3 acK49/51 acetylation. Acetylated 14-3-3 proteins are located primarily in the nucleus, where their active state has been described to bind H3 histones and many transcription factors. The inhibition of the expression of different isoforms showed that the specific silencing of the 14-3-3β gene, but not γ, increased significantly the osteogenic potential of the cells. This result correlated to the increase in acetylation of 14-3- 3β Lys 49/51 during osteogenesis. The possible role of this PTM in osteogenesis is discussed.
Collapse
Affiliation(s)
- Yesica R Frontini-López
- Laboratorio de Integración de Señales Celulares, Instituto de Histología y Embriología de Mendoza (IHEM-CONICET-UNCuyo), Mendoza, Argentina
| | - Aldana D Gojanovich
- Laboratorio de Integración de Señales Celulares, Instituto de Histología y Embriología de Mendoza (IHEM-CONICET-UNCuyo), Mendoza, Argentina.,CReM, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Samanta Del Veliz
- Laboratorio de Integración de Señales Celulares, Instituto de Histología y Embriología de Mendoza (IHEM-CONICET-UNCuyo), Mendoza, Argentina
| | - Marina Uhart
- Laboratorio de Integración de Señales Celulares, Instituto de Histología y Embriología de Mendoza (IHEM-CONICET-UNCuyo), Mendoza, Argentina
| | - Diego M Bustos
- Laboratorio de Integración de Señales Celulares, Instituto de Histología y Embriología de Mendoza (IHEM-CONICET-UNCuyo), Mendoza, Argentina.,Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
6
|
Tugaeva KV, Titterington J, Sotnikov DV, Maksimov EG, Antson AA, Sluchanko NN. Molecular basis for the recognition of steroidogenic acute regulatory protein by the 14‐3‐3 protein family. FEBS J 2020; 287:3944-3966. [DOI: 10.1111/febs.15474] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/26/2020] [Accepted: 07/01/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Kristina V. Tugaeva
- Federal Research Center of Biotechnology of the Russian Academy of Sciences A.N. Bach Institute of Biochemistry Moscow Russia
- Department of Biochemistry School of Biology M.V. Lomonosov Moscow State University Russia
| | - James Titterington
- York Structural Biology Laboratory Department of Chemistry University of York UK
| | - Dmitriy V. Sotnikov
- Federal Research Center of Biotechnology of the Russian Academy of Sciences A.N. Bach Institute of Biochemistry Moscow Russia
| | - Eugene G. Maksimov
- Federal Research Center of Biotechnology of the Russian Academy of Sciences A.N. Bach Institute of Biochemistry Moscow Russia
- Department of Biophysics School of Biology M.V. Lomonosov Moscow State University Russia
| | - Alfred A. Antson
- York Structural Biology Laboratory Department of Chemistry University of York UK
| | - Nikolai N. Sluchanko
- Federal Research Center of Biotechnology of the Russian Academy of Sciences A.N. Bach Institute of Biochemistry Moscow Russia
- Department of Biophysics School of Biology M.V. Lomonosov Moscow State University Russia
| |
Collapse
|
7
|
Chen S, Feng X, Chen X, Zhuang Z, Xiao J, Fu H, Klein JD, Wang XH, Hoover RS, Eaton DC, Cai H. 14-3-3γ, a novel regulator of the large-conductance Ca 2+-activated K + channel. Am J Physiol Renal Physiol 2020; 319:F52-F62. [PMID: 32463725 DOI: 10.1152/ajprenal.00584.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
14-3-3γ is a small protein regulating its target proteins through binding to phosphorylated serine/threonine residues. Sequence analysis of large-conductance Ca2+-activated K+ (BK) channels revealed a putative 14-3-3 binding site in the COOH-terminal region. Our previous data showed that 14-3-3γ is widely expressed in the mouse kidney. Therefore, we hypothesized that 14-3-3γ has a novel role in the regulation of BK channel activity and protein expression. We used electrophysiology, Western blot analysis, and coimmunoprecipitation to examine the effects of 14-3-3γ on BK channels both in vitro and in vivo. We demonstrated the interaction of 14-3-3γ with BK α-subunits (BKα) by coimmunoprecipitation. In human embryonic kidney-293 cells stably expressing BKα, overexpression of 14-3-3γ significantly decreased BK channel activity and channel open probability. 14-3-3γ inhibited both total and cell surface BKα protein expression while enhancing ERK1/2 phosphorylation in Cos-7 cells cotransfected with flag-14-3-3γ and myc-BK. Knockdown of 14-3-3γ by siRNA transfection markedly increased BKα expression. Blockade of the ERK1/2 pathway by incubation with the MEK-specific inhibitor U0126 partially abolished 14-3-3γ-mediated inhibition of BK protein expression. Similarly, pretreatment of the lysosomal inhibitor bafilomycin A1 reversed the inhibitory effects of 14-3-3γ on BK protein expression. Furthermore, overexpression of 14-3-3γ significantly increased BK protein ubiquitination in embryonic kidney-293 cells stably expressing BKα. Additionally, 3 days of dietary K+ challenge reduced 14-3-3γ expression and ERK1/2 phosphorylation while enhancing renal BK protein expression and K+ excretion. These data suggest that 14-3-3γ modulates BK channel activity and protein expression through an ERK1/2-mediated ubiquitin-lysosomal pathway.
Collapse
Affiliation(s)
- Shan Chen
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiuyan Feng
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Xinxin Chen
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Zhizhi Zhuang
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Jia Xiao
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Haian Fu
- Department of Pharmacology, Emory University, School of Medicine, Atlanta, Georgia
| | - Janet D Klein
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Xiaonan H Wang
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Robert S Hoover
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Section of Nephrology, Atlanta Veterans Administration Medical Center, Decatur, Georgia.,Physiology, Emory University, School of Medicine, Atlanta, Georgia
| | - Douglas C Eaton
- Physiology, Emory University, School of Medicine, Atlanta, Georgia
| | - Hui Cai
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Section of Nephrology, Atlanta Veterans Administration Medical Center, Decatur, Georgia.,Physiology, Emory University, School of Medicine, Atlanta, Georgia
| |
Collapse
|
8
|
Chung JY, Brown S, Chen H, Liu J, Papadopoulos V, Zirkin B. Effects of pharmacologically induced Leydig cell testosterone production on intratesticular testosterone and spermatogenesis†. Biol Reprod 2020; 102:489-498. [PMID: 31504200 PMCID: PMC7443349 DOI: 10.1093/biolre/ioz174] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/30/2019] [Accepted: 08/26/2019] [Indexed: 12/13/2022] Open
Abstract
The Leydig cells of the mammalian testis produce testosterone (T) in response to luteinizing hormone (LH). In rats and men with reduced serum T levels, T replacement therapy (TRT) will raise T levels, but typically with suppressive effects on sperm formation. The rate-determining step in T formation is the translocation of cholesterol to the inner mitochondrial membrane, mediated by protein-protein interactions of cytosolic and outer mitochondrial membrane proteins. Among the involved proteins is cholesterol-binding translocator protein (TSPO) (18 kDa TSPO). We hypothesized that in contrast to TRT, the administration of the TSPO agonist N,N-dihexyl-2-(4-fluorophenyl)indole-3-acetamide (FGIN-1-27), by stimulating the ability of the Leydig cells to produce T, would result in the elevation of serum T levels while maintaining intratesticular T concentration and therefore without suppression of spermatogenesis. Age-related reductions in both serum and intratesticular T levels were seen in old Brown Norway rats. Both exogenous T and FGIN-1-27 increased serum T levels. With exogenous T, serum LH and Leydig cell T formation were suppressed, and intratesticular T was reduced to below the concentration required to maintain spermatogenesis quantitatively. In contrast, FGIN-1-27 stimulated Leydig cell T formation, resulting in increased serum T without reductions in intratesticular T concentrations or in testicular sperm numbers. FGIN-1-27 also significantly increased serum and intratesticular T levels in rats made LH-deficient by treatment with the gonadotropin-releasing hormone antagonist cetrorelix. These results point to a possible approach to increasing serum T without negative effects on spermatogenesis, based upon stimulating T production by the Leydig cells themselves rather than administering T exogenously.
Collapse
Affiliation(s)
- Jin-Yong Chung
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sean Brown
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Haolin Chen
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - June Liu
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Barry Zirkin
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
9
|
Aghazadeh Y, Venugopal S, Martinez-Arguelles DB, Boisvert A, Blonder J, Papadopoulos V. Identification of Sec23ip, Part of 14-3-3γ Protein Network, as a Regulator of Acute Steroidogenesis in MA-10 Leydig Cells. Endocrinology 2020; 161:5686882. [PMID: 31875919 PMCID: PMC7007878 DOI: 10.1210/endocr/bqz036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/18/2019] [Indexed: 02/07/2023]
Abstract
Testosterone production occurs in the Leydig cells of the testes and is essential for virilization, development, reproduction, and quality of life. Although the steroidogenic proteins involved in cholesterol conversion to testosterone (T) are well characterized, the causes of reduced T during fetal, neonatal, and adult life remain uncertain. It is well established that normal cellular function is achieved through fine-tuning of multiple rather than single protein networks. Our objective was to use mass spectrometry (MS)-based proteomics to identify which cellular pathways, other than the steroidogenic machinery, influence testosterone production in MA-10 mouse tumor Leydig cells. The 14-3-3 family of scaffolds mediate protein-protein interactions facilitating the crosstalk between protein networks. We previously showed that in MA-10 cells, 14-3-3γ is a critical regulator of steroidogenesis. Therefore, identifying proteins that interact with 14-3-3γ during steroidogenesis could provide clues into the other networks involved. Using liquid chromatography (LC)-MS, we identified 688 proteins that interact with 14-3-3γ and thus potentially impact MA-10 cell steroidogenesis. The identified proteins belong to multiple protein networks, including endoplasmic reticulum-Golgi cargo sorting and vesicle biogenesis, micro ribonucleic acid-induced gene silencing, inflammation, and vesicle trafficking, to name a few. We found that silencing one of the candidates, Sec23ip, a protein known to be involved in vesicle trafficking, resulted in decreased steroidogenesis. We further showed that in Sec23ip-silenced MA-10 cells, cholesterol mobilization from the cytoplasmic membrane to mitochondria is impaired. Taken together these data suggest that Sec23ip is involved in cholesterol trafficking to supply cholesterol for acute steroidogenesis through its interactions with 14-3-3γ.
Collapse
Affiliation(s)
- Yasaman Aghazadeh
- The Research Institute of the McGill University Health Centre and the Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, Quebec, Canada
- Present address: McEwen Stem Cell Center & Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Sathvika Venugopal
- The Research Institute of the McGill University Health Centre and the Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, Quebec, Canada
| | - Daniel Benjamin Martinez-Arguelles
- The Research Institute of the McGill University Health Centre and the Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, Quebec, Canada
| | - Annie Boisvert
- The Research Institute of the McGill University Health Centre and the Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, Quebec, Canada
| | - Josip Blonder
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, sponsored by the National Cancer Institute, 8560 Progress Drive, Frederick, Maryland
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Centre and the Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, Quebec, Canada
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Ave, Los Angeles, California
- Correspondence: Vassilios Papadopoulos, Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Ave, Los Angeles, California 90089, USA. E-mail:
| |
Collapse
|
10
|
Chung JY, Chen H, Papadopoulos V, Zirkin B. Cholesterol accumulation, lipid droplet formation, and steroid production in Leydig cells: Role of translocator protein (18-kDa). Andrology 2019; 8:719-730. [PMID: 31738001 DOI: 10.1111/andr.12733] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 11/06/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cholesterol import into the mitochondria of steroid-producing cells is the rate-determining step in steroidogenesis. Numerous studies have provided evidence that the cholesterol-binding translocator protein (18 kDa TSPO) plays an important role in cholesterol translocation into mitochondria and that it also might act on cholesterol homeostasis. Several TSPO-specific ligands have been shown to increase steroid production in vitro and in vivo. OBJECTIVES The present study assessed the effects of the TSPO drug ligand FGIN-1-27 on cholesterol accumulation and lipid droplet formation in relationship to steroid formation. MATERIALS AND METHODS Using MA-10 and primary Leydig cells, immunocytochemical and molecular methods were used to examine cholesterol accumulation, the formation of lipid droplets, and steroid formation in response to LH and FGIN-1-27. Additionally, we determined the effects of Tspo knockout by CRISPR/Cas9, and of siRNA knockdowns of Tspo and Plin2 (Perilipin 2; also known as adipose differentiation-related protein, ADFP) on LH- and FGIN-1-27-induced steroidogenesis. RESULTS In response to LH and FGIN-1-27, cultured MA-10 cells and primary Leydig cells increased steroid formation, cholesterol accumulation, and lipid droplet formation. Cholesterol accumulation in the lipid droplets also was increased in Tspo knockout cells. Knockout of Tspo or its knockdown in MA-10 cells resulted in reduced progesterone formation in response to both LH and FGIN-1-27, as did knockdown of Plin2. Steroid production also was inhibited by the cholesteryl ester hydrolase inhibitor diethylumbelliferyl phosphate. DISCUSSION AND CONCLUSION These results support the conclusion that FGIN-1-27 stimulates steroid formation by increasing TSPO-mediated cholesterol translocation into the inner mitochondria for steroidogenesis, as well as into the cytosol for lipid droplet formation. FGIN-1-27 also increased steroid formation at least in part by inducing the conversion of cholesteryl ester located in lipid droplets to cholesterol, thus making available more substrate for steroid formation.
Collapse
Affiliation(s)
- Jin-Yong Chung
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Haolin Chen
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Department of Anesthesiology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Science, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Barry Zirkin
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
11
|
Tugaeva KV, Sluchanko NN. Steroidogenic Acute Regulatory Protein: Structure, Functioning, and Regulation. BIOCHEMISTRY (MOSCOW) 2019; 84:S233-S253. [PMID: 31213205 DOI: 10.1134/s0006297919140141] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Steroidogenesis takes place mainly in adrenal and gonadal cells that produce a variety of structurally similar hormones regulating numerous body functions. The rate-limiting stage of steroidogenesis is cholesterol delivery to the inner mitochondrial membrane, where it is converted by cytochrome P450scc into pregnenolone, a common precursor of all steroid hormones. The major role of supplying mitochondria with cholesterol belongs to steroidogenic acute regulatory protein (STARD1). STARD1, which is synthesized de novo as a precursor containing mitochondrial localization sequence and sterol-binding domain, significantly accelerates cholesterol transport and production of pregnenolone. Despite a tremendous interest in STARD1 fueled by its involvement in hereditary diseases and extensive efforts of numerous laboratories worldwide, many aspects of STARD1 structure, functioning, and regulation remain obscure and debatable. This review presents current concepts on the structure of STARD1 and other lipid transfer proteins, the role of STARD1 in steroidogenesis, and the mechanism of its functioning, as well as identifies the most controversial and least studied questions related to the activity of this protein.
Collapse
Affiliation(s)
- K V Tugaeva
- Bach Institute of Biochemistry, Federal Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia. .,Lomonosov Moscow State University, Biological Faculty, Department of Biochemistry, Moscow, 119234, Russia
| | - N N Sluchanko
- Bach Institute of Biochemistry, Federal Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia. .,Lomonosov Moscow State University, Biological Faculty, Department of Biophysics, Moscow, 119991, Russia
| |
Collapse
|
12
|
Kim DE, Cho CH, Sim KM, Kwon O, Hwang EM, Kim HW, Park JY. 14-3-3γ Haploinsufficient Mice Display Hyperactive and Stress-sensitive Behaviors. Exp Neurobiol 2019; 28:43-53. [PMID: 30853823 PMCID: PMC6401549 DOI: 10.5607/en.2019.28.1.43] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/16/2019] [Accepted: 01/18/2019] [Indexed: 02/03/2023] Open
Abstract
14-3-3γ plays diverse roles in different aspects of cellular processes. Especially in the brain where 14-3-3γ is enriched, it has been reported to be involved in neurological and psychiatric diseases (e.g. Williams-Beuren syndrome and Creutzfeldt-Jakob disease). However, behavioral abnormalities related to 14-3-3γ deficiency are largely unknown. Here, by using 14-3-3γ deficient mice, we found that homozygous knockout mice were prenatally lethal, and heterozygous mice showed developmental delay relative to wild-type littermate mice. In addition, in behavioral analyses, we found that 14-3-3γ heterozygote mice display hyperactive and depressive-like behavior along with more sensitive responses to acute stress than littermate control mice. These results suggest that 14-3-3γ levels may be involved in the developmental manifestation of related neuropsychiatric diseases. In addition, 14-3-3γ heterozygote mice may be a potential model to study the molecular pathophysiology of neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Do Eon Kim
- College of Life Sciences, Sejong University, Seoul 05006, Korea
| | - Chang-Hoon Cho
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Kyoung Mi Sim
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Osung Kwon
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Hyung-Wook Kim
- College of Life Sciences, Sejong University, Seoul 05006, Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| |
Collapse
|
13
|
Pennington KL, Chan TY, Torres MP, Andersen JL. The dynamic and stress-adaptive signaling hub of 14-3-3: emerging mechanisms of regulation and context-dependent protein-protein interactions. Oncogene 2018; 37:5587-5604. [PMID: 29915393 PMCID: PMC6193947 DOI: 10.1038/s41388-018-0348-3] [Citation(s) in RCA: 240] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/07/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
14-3-3 proteins are a family of structurally similar phospho-binding proteins that regulate essentially every major cellular function. Decades of research on 14-3-3s have revealed a remarkable network of interacting proteins that demonstrate how 14-3-3s integrate and control multiple signaling pathways. In particular, these interactions place 14-3-3 at the center of the signaling hub that governs critical processes in cancer, including apoptosis, cell cycle progression, autophagy, glucose metabolism, and cell motility. Historically, the majority of 14-3-3 interactions have been identified and studied under nutrient-replete cell culture conditions, which has revealed important nutrient driven interactions. However, this underestimates the reach of 14-3-3s. Indeed, the loss of nutrients, growth factors, or changes in other environmental conditions (e.g., genotoxic stress) will not only lead to the loss of homeostatic 14-3-3 interactions, but also trigger new interactions, many of which are likely stress adaptive. This dynamic nature of the 14-3-3 interactome is beginning to come into focus as advancements in mass spectrometry are helping to probe deeper and identify context-dependent 14-3-3 interactions-providing a window into adaptive phosphorylation-driven cellular mechanisms that orchestrate the tumor cell's response to a variety of environmental conditions including hypoxia and chemotherapy. In this review, we discuss emerging 14-3-3 regulatory mechanisms with a focus on post-translational regulation of 14-3-3 and dynamic protein-protein interactions that illustrate 14-3-3's role as a stress-adaptive signaling hub in cancer.
Collapse
Affiliation(s)
- K L Pennington
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - T Y Chan
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - M P Torres
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - J L Andersen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA.
| |
Collapse
|
14
|
Gadahi JA, Wang S, Bo G, Ehsan M, Yan R, Song X, Xu L, Li X. Proteomic Analysis of the Excretory and Secretory Proteins of Haemonchus contortus (HcESP) Binding to Goat PBMCs In Vivo Revealed Stage-Specific Binding Profiles. PLoS One 2016; 11:e0159796. [PMID: 27467391 PMCID: PMC4965049 DOI: 10.1371/journal.pone.0159796] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 07/09/2016] [Indexed: 02/06/2023] Open
Abstract
Haemonchus contortus is a parasitic gastrointestinal nematode, and its excretory and secretory products (HcESPs) interact extensively with the host cells. In this study, we report the interaction of proteins from HcESPs at different developmental stages to goat peripheral blood mononuclear cells (PBMCs) in vivo using liquid chromatography-tandem mass spectrometry. A total of 407 HcESPs that interacted with goat PBMCs at different time points were identified from a H. contortus protein database using SEQUEST searches. The L4 and L5 stages of H. contortus represented a higher proportion of the identified proteins compared with the early and late adult stages. Both stage-specific interacting proteins and proteins that were common to multiple stages were identified. Forty-seven interacting proteins were shared among all stages. The gene ontology (GO) distributions of the identified goat PBMC-interacting proteins were nearly identical among all developmental stages, with high representation of binding and catalytic activity. Cellular, metabolic and single-organism processes were also annotated as major biological processes, but interestingly, more proteins were annotated as localization processes at the L5 stage than at the L4 and adult stages. Based on the clustering of homologous proteins, we improved the functional annotations of un-annotated proteins identified at different developmental stages. Some unnamed H. contortus ATP-binding cassette proteins, including ADP-ribosylation factor and P-glycoprotein-9, were identified by STRING protein clustering analysis.
Collapse
Affiliation(s)
- Javaid Ali Gadahi
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Shuai Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Gao Bo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Muhammad Ehsan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - RuoFeng Yan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - XiaoKai Song
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - LiXin Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - XiangRui Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| |
Collapse
|
15
|
Sterol Carrier Protein-2, a Nonspecific Lipid-Transfer Protein, in Intracellular Cholesterol Trafficking in Testicular Leydig Cells. PLoS One 2016; 11:e0149728. [PMID: 26901662 PMCID: PMC4762939 DOI: 10.1371/journal.pone.0149728] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 01/26/2016] [Indexed: 11/19/2022] Open
Abstract
Sterol carrier protein-2 (SCP2), also called nonspecific lipid-transfer protein, is thought to play a major role in intracellular lipid transport and metabolism, and it has been associated with diseases involving abnormalities in lipid trafficking, such as Zellweger syndrome. The Scp2 gene encodes the 58 kDa sterol carrier protein-x (SCPX) and 15 kDa pro-SCP2 proteins, both of which contain a 13 kDa SCP2 domain in their C-termini. We found that 22-NBD-cholesterol, a fluorescent analog of cholesterol and a preferred SCP2 ligands, was not localized in the peroxisomes. This raises questions about previous reports on the localization of the SCPX and SCP2 proteins and their relationship to peroxisomes and mitochondria in intracellular cholesterol transport. Immunofluorescent staining of cryosections of mouse testis and of MA-10 mouse tumor Leydig cells showed that SCPX and SCP2 are present in both mouse testicular interstitial tissue and in MA-10 cells. Fluorescent fusion proteins of SCPX and SCP2, as well as confocal live-cell imaging, were used to investigate the subcellular targeting of these proteins and the function of the putative mitochondrial targeting sequence. The results showed that SCPX and SCP2 are targeted to the peroxisomes by the C-terminal PTS1 domain, but the putative N-terminal mitochondrial targeting sequence alone is not potent enough to localize SCPX and SCP2 to the mitochondria. Homology modeling and molecular docking studies indicated that the SCP2 domain binds cholesterol, but lacks specificity of the binding and/or transport. These findings further our understanding of the role of SCPX and SCP2 in intracellular cholesterol transport, and present a new point of view on the role of these proteins in cholesterol trafficking.
Collapse
|
16
|
Graham A. Mitochondrial regulation of macrophage cholesterol homeostasis. Free Radic Biol Med 2015; 89:982-92. [PMID: 26416507 DOI: 10.1016/j.freeradbiomed.2015.08.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/28/2015] [Accepted: 08/11/2015] [Indexed: 12/19/2022]
Abstract
This review explores the relationship between mitochondrial structure and function in the regulation of macrophage cholesterol metabolism and proposes that mitochondrial dysfunction contributes to loss of the elegant homeostatic mechanisms which normally maintain cellular sterol levels within defined limits. Mitochondrial sterol 27-hydroxylase (CYP27A1) can generate oxysterol activators of liver X receptors which heterodimerise with retinoid X receptors, enhancing the transcription of ATP binding cassette transporters (ABCA1, ABCG1, and ABCG4), that can remove excess cholesterol via efflux to apolipoproteins A-1, E, and high density lipoprotein, and inhibit inflammation. The activity of CYP27A1 is regulated by the rate of supply of cholesterol substrate to the inner mitochondrial membrane, mediated by a complex of proteins. The precise identity of this dynamic complex remains controversial, even in steroidogenic tissues, but may include steroidogenic acute regulatory protein and the 18 kDa translocator protein, together with voltage-dependent anion channels, ATPase AAA domain containing protein 3A, and optic atrophy type 1 proteins. Certainly, overexpression of StAR and TSPO proteins can enhance macrophage cholesterol efflux to apoA-I and/or HDL, while perturbations in mitochondrial function, or changes in the expression of mitochondrial fusion proteins, alter the efficiency of cholesterol efflux. Molecules which can sustain or improve mitochondrial function or increase the activity of the protein complex involved in cholesterol transfer may have utility in resolving the problem of dysregulated macrophage cholesterol homeostasis, a condition which may contribute to inflammation, atherosclerosis, nonalcoholic steatohepatitis, osteoblastic bone resorption, and some disorders of the central nervous system.
Collapse
Affiliation(s)
- Annette Graham
- Department of Life Sciences, School of Health and Life Sciences, and Institute for Applied Health Research, Glasgow Caledonian University, 70 Cowcaddens Road, Glasgow G4 0BA, United Kingdom.
| |
Collapse
|
17
|
Aghazadeh Y, Papadopoulos V. The role of the 14-3-3 protein family in health, disease, and drug development. Drug Discov Today 2015; 21:278-87. [PMID: 26456530 DOI: 10.1016/j.drudis.2015.09.012] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 09/21/2015] [Accepted: 09/29/2015] [Indexed: 11/18/2022]
Abstract
14-3-3 proteins regulate intracellular signaling pathways, such as signal transduction, protein trafficking, cell cycle, and apoptosis. In addition to the ubiquitous roles of 14-3-3 isoforms, unique tissue-specific functions are also described for each isoform. Owing to their role in regulating cell cycle, protein trafficking, and steroidogenesis, 14-3-3 proteins are prevalent in human diseases, such as cancer, neurodegeneration, and reproductive disorders, and, therefore, serve as valuable drug targets. In this review, we summarize the role of 14-3-3 proteins in normal and disease states, with a focus on 14-3-3γ and ɛ. We also discuss drug compounds targeting 14-3-3 proteins and their potential therapeutic uses.
Collapse
Affiliation(s)
- Yasaman Aghazadeh
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; Department of Medicine, McGill University, Montreal, QC H3G 1A4, Canada
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; Department of Medicine, McGill University, Montreal, QC H3G 1A4, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
18
|
Sluchanko NN, Uversky VN. Hidden disorder propensity of the N-terminal segment of universal adapter protein 14-3-3 is manifested in its monomeric form: Novel insights into protein dimerization and multifunctionality. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:492-504. [PMID: 25747569 DOI: 10.1016/j.bbapap.2015.02.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 02/17/2015] [Accepted: 02/24/2015] [Indexed: 11/30/2022]
Abstract
The multiplicity of functions of 14-3-3 proteins, integrated into many cellular interactions and signaling networks, is primarily based upon their dimeric α-helical structure that is capable of binding phosphorylated protein partners as well as displaying a "moonlighting" chaperone-like activity. The structure and functions of 14-3-3 proteins are regulated in different ways, including Ser58 phosphorylation in the interface, which shifts equilibrium towards the formation of protein monomers whose role is poorly understood. While modification of Ser58 induced only partial dissociation, the engineered triple mutation of human 14-3-3ζ located in the first α-helix deeply monomerized the protein, allowing for a structural analysis of the monomeric form. Dimer-incapable 14-3-3 proteins retained binding capacity and specificity towards some phosphopartners, and also demonstrated increased chaperone-like activity on various substrates. Here, we found a substantial propensity of the N-terminal segment (~40 residues) of 14-3-3 proteins to intrinsic disorder, showing remarkable conservation across different isoforms and organisms. We hypothesized that this intrinsic disorder propensity, hidden in the α-helical 14-3-3 dimer, can be manifested upon its dissociation and interrogated novel monomeric 14-3-3ζ carrying both monomerizing and S58E mutations (14-3-3ζmS58E). CD spectroscopy showed that, at physiological temperatures, this protein has ~10-15% reduced helicity relative to the wild type protein, corresponding to roughly 40 residues. Along with the known flexibility of C-terminus, SAXS-based modeling of the 14-3-3ζmS58E structure strongly suggested pliability of its N-terminus. The unraveled disorder propensity of the N-terminal tails of 14-3-3 proteins provides new clues for better understanding of the molecular mechanisms of dimerization and multifunctionality of these universal adapter proteins.
Collapse
Affiliation(s)
- Nikolai N Sluchanko
- A. N. Bach Institute of Biochemistry, Russian Academy of Sciences, Leninsky prospect 33, Moscow 119071, Russian Federation.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow Region, Russian Federation; Biology Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russian Federation.
| |
Collapse
|