1
|
Gao X, Sun Y, Huang X, Zhou Y, Zhu H, Li Q, Ma Y. Adequate dietary magnesium intake may protect females but not males older than 55 years from cognitive impairment. Nutr Neurosci 2024; 27:184-195. [PMID: 36803323 DOI: 10.1080/1028415x.2023.2169986] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
BACKGROUND Magnesium is an essential nutrient required to maintain brain health throughout life, and adequate magnesium intake is positively associated with cognitive performance in older adults. However, sex differences in magnesium metabolism have not been adequately assessed in humans. OBJECTIVES We investigated sex differences in the effect of dietary magnesium intake and the risk of different types of cognitive impairment in older Chinese adults. METHODS We collected and assessed dietary data and cognitive function status in people aged 55 years and older in northern China who participated in the Community Cohort Study of Nervous System Diseases from 2018 to 2019 to explore the relationship between dietary magnesium intake and the risk of each type of mild cognitive impairment (MCI) in sex-specific cohorts of older adults. RESULTS The study included 612 people: 260 (42.5%) men and 352 (57.5%) women. Logistic regression results showed that for the total sample and women's sample, high dietary magnesium intake reduced the risk of amnestic MCI (ORtotal = 0.300; ORwomen = 0.190) and multidomain amnestic MCI (ORtotal = 0.225; ORwomen = 0.145). The results of restricted cubic spline analysis showed that the risk of amnestic MCI (ptotal = 0.0193; pwomen = 0.0351) and multidomain amnestic MCI (ptotal = 0.0089; pwomen = 0.0096) in the total sample and women's sample gradually decreased with increasing dietary magnesium intake. CONCLUSIONS The results suggest that adequate magnesium intake may have a preventive effect against the risk of MCI in older women.
Collapse
Affiliation(s)
- Xian Gao
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, People's Republic of China
- Department of Gastrointestinal Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yan Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, People's Republic of China
| | - Xin Huang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, People's Republic of China
- Handan Center for Disease Control and Prevention, Handan, People's Republic of China
| | - Yutian Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, People's Republic of China
| | - Huichen Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, People's Republic of China
| | - Qingxia Li
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, People's Republic of China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, People's Republic of China
| |
Collapse
|
2
|
Kholodenko IV, Kholodenko RV, Yarygin KN. The Crosstalk between Mesenchymal Stromal/Stem Cells and Hepatocytes in Homeostasis and under Stress. Int J Mol Sci 2023; 24:15212. [PMID: 37894893 PMCID: PMC10607347 DOI: 10.3390/ijms242015212] [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: 09/23/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Liver diseases, characterized by high morbidity and mortality, represent a substantial medical problem globally. The current therapeutic approaches are mainly aimed at reducing symptoms and slowing down the progression of the diseases. Organ transplantation remains the only effective treatment method in cases of severe liver pathology. In this regard, the development of new effective approaches aimed at stimulating liver regeneration, both by activation of the organ's own resources or by different therapeutic agents that trigger regeneration, does not cease to be relevant. To date, many systematic reviews and meta-analyses have been published confirming the effectiveness of mesenchymal stromal cell (MSC) transplantation in the treatment of liver diseases of various severities and etiologies. However, despite the successful use of MSCs in clinical practice and the promising therapeutic results in animal models of liver diseases, the mechanisms of their protective and regenerative action remain poorly understood. Specifically, data about the molecular agents produced by these cells and mediating their therapeutic action are fragmentary and often contradictory. Since MSCs or MSC-like cells are found in all tissues and organs, it is likely that many key intercellular interactions within the tissue niches are dependent on MSCs. In this context, it is essential to understand the mechanisms underlying communication between MSCs and differentiated parenchymal cells of each particular tissue. This is important both from the perspective of basic science and for the development of therapeutic approaches involving the modulation of the activity of resident MSCs. With regard to the liver, the research is concentrated on the intercommunication between MSCs and hepatocytes under normal conditions and during the development of the pathological process. The goals of this review were to identify the key factors mediating the crosstalk between MSCs and hepatocytes and determine the possible mechanisms of interaction of the two cell types under normal and stressful conditions. The analysis of the hepatocyte-MSC interaction showed that MSCs carry out chaperone-like functions, including the synthesis of the supportive extracellular matrix proteins; prevention of apoptosis, pyroptosis, and ferroptosis; support of regeneration; elimination of lipotoxicity and ER stress; promotion of antioxidant effects; and donation of mitochondria. The underlying mechanisms suggest very close interdependence, including even direct cytoplasm and organelle exchange.
Collapse
Affiliation(s)
- Irina V. Kholodenko
- Laboratory of Cell Biology, Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - Roman V. Kholodenko
- Laboratory of Molecular Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia;
| | - Konstantin N. Yarygin
- Laboratory of Cell Biology, Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| |
Collapse
|
3
|
Hardy S, Zolotarov Y, Coleman J, Roitman S, Khursheed H, Aubry I, Uetani N, Tremblay M. PRL-1/2 phosphatases control TRPM7 magnesium-dependent function to regulate cellular bioenergetics. Proc Natl Acad Sci U S A 2023; 120:e2221083120. [PMID: 36972446 PMCID: PMC10083557 DOI: 10.1073/pnas.2221083120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/03/2023] [Indexed: 03/29/2023] Open
Abstract
Phosphatases of regenerating liver (PRL-1, PRL-2, PRL-3; also known as PTP4A1, PTP4A2, PTP4A3, respectively) control intracellular magnesium levels by interacting with the CNNM magnesium transport regulators. Still, the exact mechanism governing magnesium transport by this protein complex is not well understood. Herein, we have developed a genetically encoded intracellular magnesium-specific reporter and demonstrate that the CNNM family inhibits the function of the TRPM7 magnesium channel. We show that the small GTPase ARL15 increases CNNM3/TRPM7 protein complex formation to reduce TRPM7 activity. Conversely, PRL-2 overexpression counteracts ARL15 binding to CNNM3 and enhances the function of TRPM7 by preventing the interaction between CNNM3 and TRPM7. Moreover, while TRPM7-induced cell signaling is promoted by PRL-1/2, it is reduced when CNNM3 is overexpressed. Lowering cellular magnesium levels reduces the interaction of CNNM3 with TRPM7 in a PRL-dependent manner, whereby knockdown of PRL-1/2 restores the protein complex formation. Cotargeting of TRPM7 and PRL-1/2 alters mitochondrial function and sensitizes cells to metabolic stress induced by magnesium depletion. These findings reveal the dynamic regulation of TRPM7 function in response to PRL-1/2 levels, to coordinate magnesium transport and reprogram cellular metabolism.
Collapse
Affiliation(s)
- Serge Hardy
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Yevgen Zolotarov
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Jacob Coleman
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Simon Roitman
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Hira Khursheed
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Isabelle Aubry
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Noriko Uetani
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| | - Michel L. Tremblay
- Goodman Cancer Institute, McGill University, Montreal, QCH3A1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QCH3A1A3, Canada
| |
Collapse
|
4
|
Kim JY, Kim SH, Seok J, Bae SH, Hwang SG, Kim GJ. Increased PRL-1 in BM-derived MSCs triggers anaerobic metabolism via mitochondria in a cholestatic rat model. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:512-524. [PMID: 36865088 PMCID: PMC9970868 DOI: 10.1016/j.omtn.2023.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 01/31/2023] [Indexed: 02/09/2023]
Abstract
Mesenchymal stem cell (MSC) therapy in chronic liver disease is associated with mitochondrial anaerobic metabolism. Phosphatase of regenerating liver-1 (PRL-1), known as protein tyrosine phosphatase type 4A, member 1 (PTP4A1), plays a critical role in liver regeneration. However, its therapeutic mechanism remains obscure. The aim of this study was to establish genetically modified bone marrow (BM)-MSCs overexpressing PRL-1 (BM-MSCsPRL-1) and to investigate their therapeutic effects on mitochondrial anaerobic metabolism in a bile duct ligation (BDL)-injured cholestatic rat model. BM-MSCsPRL-1 were generated with lentiviral and nonviral gene delivery systems and characterized. Compared with naive cells, BM-MSCsPRL-1 showed an improved antioxidant capacity and mitochondrial dynamics and decreased cellular senescence. In particular, mitochondrial respiration in BM-MSCsPRL-1 generated using the nonviral system was significantly increased as well as mtDNA copy number and total ATP production. Moreover, transplantation of BM-MSCsPRL-1 generated using the nonviral system had predominantly antifibrotic effects and restored hepatic function in a BDL rat model. Decreased cytoplasmic lactate and increased mitochondrial lactate upon the administration of BM-MSCsPRL-1 indicated significant alterations in mtDNA copy number and ATP production, activating anaerobic metabolism. In conclusion, BM-MSCsPRL-1 generated by a nonviral gene delivery system enhanced anaerobic mitochondrial metabolism in a cholestatic rat model, improving hepatic function.
Collapse
Affiliation(s)
- Jae Yeon Kim
- Department of Biomedical Science, CHA University, 689, Sampyeong-dong, Bundang-gu, Seongnam-si 13488, Republic of Korea,Research Institute of Placental Science, CHA University, Seongnam 13488, Republic of Korea
| | - Se Ho Kim
- Department of Biomedical Science, CHA University, 689, Sampyeong-dong, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Jin Seok
- Department of Biomedical Science, CHA University, 689, Sampyeong-dong, Bundang-gu, Seongnam-si 13488, Republic of Korea,Research Institute of Placental Science, CHA University, Seongnam 13488, Republic of Korea
| | - Si Hyun Bae
- Department of Internal Medicine, Catholic University Medical College, Seoul 03312, Republic of Korea
| | - Seong-Gyu Hwang
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam 13496, Republic of Korea
| | - Gi Jin Kim
- Department of Biomedical Science, CHA University, 689, Sampyeong-dong, Bundang-gu, Seongnam-si 13488, Republic of Korea,Research Institute of Placental Science, CHA University, Seongnam 13488, Republic of Korea,Corresponding author Gi Jin Kim, Department of Biomedical Science, CHA University, 689, Sampyeong-dong, Bundang-gu, Seongnam-si 13488, Republic of Korea.
| |
Collapse
|
5
|
Chronoradiobiology of Breast Cancer: The Time Is Now to Link Circadian Rhythm and Radiation Biology. Int J Mol Sci 2022; 23:ijms23031331. [PMID: 35163264 PMCID: PMC8836288 DOI: 10.3390/ijms23031331] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/23/2022] [Indexed: 12/13/2022] Open
Abstract
Circadian disruption has been linked to cancer development, progression, and radiation response. Clinical evidence to date shows that circadian genetic variation and time of treatment affect radiation response and toxicity for women with breast cancer. At the molecular level, there is interplay between circadian clock regulators such as PER1, which mediates ATM and p53-mediated cell cycle gating and apoptosis. These molecular alterations may govern aggressive cancer phenotypes, outcomes, and radiation response. Exploiting the various circadian clock mechanisms may enhance the therapeutic index of radiation by decreasing toxicity, increasing disease control, and improving outcomes. We will review the body’s natural circadian rhythms and clock gene-regulation while exploring preclinical and clinical evidence that implicates chronobiological disruptions in the etiology of breast cancer. We will discuss radiobiological principles and the circadian regulation of DNA damage responses. Lastly, we will present potential rational therapeutic approaches that target circadian pathways to improve outcomes in breast cancer. Understanding the implications of optimal timing in cancer treatment and exploring ways to entrain circadian biology with light, diet, and chronobiological agents like melatonin may provide an avenue for enhancing the therapeutic index of radiotherapy.
Collapse
|
6
|
Gehring K, Kozlov G, Yang M, Fakih R. The double lives of phosphatases of regenerating liver: A structural view of their catalytic and noncatalytic activities. J Biol Chem 2021; 298:101471. [PMID: 34890645 PMCID: PMC8728433 DOI: 10.1016/j.jbc.2021.101471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 11/18/2022] Open
Abstract
Phosphatases of regenerating liver (PRLs) are protein phosphatases involved in the control of cell growth and migration. They are known to promote cancer metastasis but, despite over 20 years of study, there is still no consensus about their mechanism of action. Recent work has revealed that PRLs lead double lives, acting both as catalytically active enzymes and as pseudophosphatases. The three known PRLs belong to the large family of cysteine phosphatases that form a phosphocysteine intermediate during catalysis. Uniquely to PRLs, this intermediate is stable, with a lifetime measured in hours. As a consequence, PRLs have very little phosphatase activity. Independently, PRLs also act as pseudophosphatases by binding CNNM membrane proteins to regulate magnesium homeostasis. In this function, an aspartic acid from CNNM inserts into the phosphatase catalytic site of PRLs, mimicking a substrate–enzyme interaction. The delineation of PRL pseudophosphatase and phosphatase activities in vivo was impossible until the recent identification of PRL mutants defective in one activity or the other. These mutants showed that CNNM binding was sufficient for PRL oncogenicity in one model of metastasis, but left unresolved its role in other contexts. As the presence of phosphocysteine prevents CNNM binding and CNNM-binding blocks catalytic activity, these two activities are inherently linked. Additional studies are needed to untangle the intertwined catalytic and noncatalytic functions of PRLs. Here, we review the current understanding of the structure and biophysical properties of PRL phosphatases.
Collapse
Affiliation(s)
- Kalle Gehring
- Department of Biochemistry and Centre de Recherche en Biologie Structurale, McGill University, Montreal, Quebec, Canada.
| | - Guennadi Kozlov
- Department of Biochemistry and Centre de Recherche en Biologie Structurale, McGill University, Montreal, Quebec, Canada
| | - Meng Yang
- Department of Biochemistry and Centre de Recherche en Biologie Structurale, McGill University, Montreal, Quebec, Canada
| | - Rayan Fakih
- Department of Biochemistry and Centre de Recherche en Biologie Structurale, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
7
|
ARL15 modulates magnesium homeostasis through N-glycosylation of CNNMs. Cell Mol Life Sci 2021; 78:5427-5445. [PMID: 34089346 PMCID: PMC8257531 DOI: 10.1007/s00018-021-03832-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 03/23/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023]
Abstract
Cyclin M (CNNM1-4) proteins maintain cellular and body magnesium (Mg2+) homeostasis. Using various biochemical approaches, we have identified members of the CNNM family as direct interacting partners of ADP-ribosylation factor-like GTPase 15 (ARL15), a small GTP-binding protein. ARL15 interacts with CNNMs at their carboxyl-terminal conserved cystathionine-β-synthase (CBS) domains. In silico modeling of the interaction between CNNM2 and ARL15 supports that the small GTPase specifically binds the CBS1 and CNBH domains. Immunocytochemical experiments demonstrate that CNNM2 and ARL15 co-localize in the kidney, with both proteins showing subcellular localization in the endoplasmic reticulum, Golgi apparatus and the plasma membrane. Most importantly, we found that ARL15 is required for forming complex N-glycosylation of CNNMs. Overexpression of ARL15 promotes complex N-glycosylation of CNNM3. Mg2+ uptake experiments with a stable isotope demonstrate that there is a significant increase of 25Mg2+ uptake upon knockdown of ARL15 in multiple kidney cancer cell lines. Altogether, our results establish ARL15 as a novel negative regulator of Mg2+ transport by promoting the complex N-glycosylation of CNNMs.
Collapse
|
8
|
PRL-2 phosphatase is required for vascular morphogenesis and angiogenic signaling. Commun Biol 2020; 3:603. [PMID: 33097786 PMCID: PMC7584612 DOI: 10.1038/s42003-020-01343-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 09/30/2020] [Indexed: 12/21/2022] Open
Abstract
Protein tyrosine phosphatases are essential modulators of angiogenesis and have been identified as novel therapeutic targets in cancer and anti-angiogenesis. The roles of atypical Phosphatase of Regenerative Liver (PRL) phosphatases in this context remain poorly understood. Here, we investigate the biological function of PRL phosphatases in developmental angiogenesis in the postnatal mouse retina and in cell culture. We show that endothelial cells in the retina express PRL-2 encoded by the Ptp4a2 gene, and that inducible endothelial and global Ptp4a2 mutant mice exhibit defective retinal vascular outgrowth, arteriovenous differentiation, and sprouting angiogenesis. Mechanistically, PTP4A2 deletion limits angiogenesis by inhibiting endothelial cell migration and the VEGF-A, DLL-4/NOTCH-1 signaling pathway. This study reveals the importance of PRL-2 as a modulator of vascular development.
Collapse
|
9
|
Winiger EA, Huggett SB, Hatoum AS, Stallings MC, Hewitt JK. Onset of regular cannabis use and adult sleep duration: Genetic variation and the implications of a predictive relationship. Drug Alcohol Depend 2019; 204:107517. [PMID: 31698253 PMCID: PMC7053256 DOI: 10.1016/j.drugalcdep.2019.06.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/14/2019] [Accepted: 06/19/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Limited evidence suggests that early cannabis use is associated with sleep problems. Research is needed to understand the developmental impact of early regular cannabis use on later adult sleep duration. METHODS In a sample of 1656 adult twins (56% female, Mean age = 25.79yrs), linear mixed effects models were used to analyze the influence of retrospectively assessed age of onset of regular cannabis use on adult sleep duration controlling for sex, depression, and current substance use. Twin analyses provided genetic and environmental variance estimates as well as insights into the association and potential casual relationships between these traits. RESULTS Earlier age of onset for regular cannabis use was significantly associated with shorter adult sleep duration on both weekdays (β = -0.13, 95% CI = [-0.23, -0.04]) and weekends (β = -0.18, 95% CI = [-0.27, -0.08]). Additive genetics significantly contributed to the onset of regular cannabis use (a2 = 76%, 95% CI = [68, 85]) and adult weekend sleep duration (a2 = 20%, 95% CI = [11, 32]). We found evidence of a significant genetic correlation (rA = -0.31, 95% CI = [-0.41, -0.15]) between these two traits and our best fitting model was consistent with early onset of regular cannabis use causing shorter adult weekend sleep duration (β = -0.11, 95% CI = [-0.18, -0.03]). CONCLUSIONS Our results are consistent with the hypothesis that early onset of regular cannabis use may have a negative impact on adult sleep duration.
Collapse
Affiliation(s)
- Evan A. Winiger
- Institute for Behavioral Genetics, University of Colorado, Boulder, East Campus, 1480 30th Street, Boulder, CO 80309, United States,Department of Psychology and Neuroscience, University of Colorado, Boulder, Muenzinger Psychology Building, 1905 Colorado Ave, Boulder, CO 80309, United States,Corresponding author at: Institute for Behavioral Genetics, University of Colorado, Boulder, Boulder, CO 80309, United States. (E.A. Winiger)
| | - Spencer B. Huggett
- Institute for Behavioral Genetics, University of Colorado, Boulder, East Campus, 1480 30th Street, Boulder, CO 80309, United States,Department of Psychology and Neuroscience, University of Colorado, Boulder, Muenzinger Psychology Building, 1905 Colorado Ave, Boulder, CO 80309, United States
| | - Alexander S. Hatoum
- Institute for Behavioral Genetics, University of Colorado, Boulder, East Campus, 1480 30th Street, Boulder, CO 80309, United States,Department of Psychology and Neuroscience, University of Colorado, Boulder, Muenzinger Psychology Building, 1905 Colorado Ave, Boulder, CO 80309, United States
| | - Michael C. Stallings
- Institute for Behavioral Genetics, University of Colorado, Boulder, East Campus, 1480 30th Street, Boulder, CO 80309, United States,Department of Psychology and Neuroscience, University of Colorado, Boulder, Muenzinger Psychology Building, 1905 Colorado Ave, Boulder, CO 80309, United States
| | - John K. Hewitt
- Institute for Behavioral Genetics, University of Colorado, Boulder, East Campus, 1480 30th Street, Boulder, CO 80309, United States,Department of Psychology and Neuroscience, University of Colorado, Boulder, Muenzinger Psychology Building, 1905 Colorado Ave, Boulder, CO 80309, United States
| |
Collapse
|
10
|
Kanellopoulou C, George AB, Masutani E, Cannons JL, Ravell JC, Yamamoto TN, Smelkinson MG, Jiang PD, Matsuda-Lennikov M, Reilley J, Handon R, Lee PH, Miller JR, Restifo NP, Zheng L, Schwartzberg PL, Young M, Lenardo MJ. Mg 2+ regulation of kinase signaling and immune function. J Exp Med 2019; 216:1828-1842. [PMID: 31196981 PMCID: PMC6683994 DOI: 10.1084/jem.20181970] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/22/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
A Mg2+-dependent mechanism regulates proximal T cell receptor signaling by modulating ITK activity through a low-affinity Mg2+ binding pocket in the catalytic domain. Dietary Mg2+ deprivation in mice impairs T cell activation and T cell–mediated immunity against influenza. Mg2+ is required at micromolar concentrations as a cofactor for ATP, enzymatic reactions, and other biological processes. We show that decreased extracellular Mg2+ reduced intracellular Mg2+ levels and impaired the Ca2+ flux, activation marker up-regulation, and proliferation after T cell receptor (TCR) stimulation. Reduced Mg2+ specifically impairs TCR signal transduction by IL-2–inducible T cell kinase (ITK) due to a requirement for a regulatory Mg2+ in the catalytic pocket of ITK. We also show that altered catalytic efficiency by millimolar changes in free basal Mg2+ is an unrecognized but conserved feature of other serine/threonine and tyrosine kinases, suggesting a Mg2+ regulatory paradigm of kinase function. Finally, a reduced serum Mg2+ concentration in mice causes an impaired CD8+ T cell response to influenza A virus infection, reduces T cell activation, and exacerbates morbidity. Thus, Mg2+ directly regulates the active site of specific kinases during T cell responses, and maintaining a high serum Mg2+ concentration is important for antiviral immunity in otherwise healthy animals.
Collapse
Affiliation(s)
- Chryssa Kanellopoulou
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Alex B George
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Evan Masutani
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Medical Scientist Training Program, School of Medicine, University of California, San Diego, San Diego, CA
| | - Jennifer L Cannons
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Genetic Disease Research Branch, National Human Genome Research Institute, Bethesda, MD
| | - Juan C Ravell
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Tori N Yamamoto
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.,Center for Cell-Based Therapy, National Cancer Institute, National Institutes of Health, Bethesda, MD.,Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA
| | - Margery G Smelkinson
- Biological Imaging, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Ping Du Jiang
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Mami Matsuda-Lennikov
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Julie Reilley
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Genetic Disease Research Branch, National Human Genome Research Institute, Bethesda, MD
| | - Robin Handon
- Genetic Disease Research Branch, National Human Genome Research Institute, Bethesda, MD
| | - Ping-Hsien Lee
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.,Center for Cell-Based Therapy, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | | | - Nicholas P Restifo
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.,Center for Cell-Based Therapy, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lixin Zheng
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Pamela L Schwartzberg
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Genetic Disease Research Branch, National Human Genome Research Institute, Bethesda, MD
| | - Matthew Young
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Michael J Lenardo
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD .,Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
11
|
Zhang Z, Kozlov G, Chen YS, Gehring K. Mechanism of thienopyridone and iminothienopyridinedione inhibition of protein phosphatases. MEDCHEMCOMM 2019; 10:791-799. [PMID: 31191869 DOI: 10.1039/c9md00175a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 04/02/2019] [Indexed: 12/14/2022]
Abstract
Thienopyridone (TP) has been proposed as a selective inhibitor of phosphatases of regenerating liver (PRL or PTP4A). PRLs are dual specificity phosphatases that promote cancer progression and are attractive anticancer targets. TP and iminothienopyridinedione (ITP), a more potent derivative, were shown to be effective inhibitors but the mechanism of inhibition was not established. Here, we perform NMR experiments and in vitro phosphatase assays to show that TP and ITP inhibit protein phosphatases non-specifically through oxidation of the phosphatase catalytic cysteine. We demonstrate that TP and ITP are redox active compounds, inhibiting PRL-3 and multiple other PTPs through oxidation. They also catalyze the oxidation of thioredoxin-1 as well as small molecules, like TCEP, DTT, and glutathione. The reported selectivity of TP and ITP is likely due to the higher susceptibility of PRLs to oxidation. Thus, while TP and ITP effectively inhibit PRLs, their use for studying the cellular function of PRLs is problematic due to the likelihood of off-target effects.
Collapse
Affiliation(s)
- Zhidian Zhang
- Department of Biochemistry and Centre for Structural Biology , McGill University , Montreal , Quebec , Canada .
| | - Guennadi Kozlov
- Department of Biochemistry and Centre for Structural Biology , McGill University , Montreal , Quebec , Canada .
| | - Yu Seby Chen
- Department of Biochemistry and Centre for Structural Biology , McGill University , Montreal , Quebec , Canada .
| | - Kalle Gehring
- Department of Biochemistry and Centre for Structural Biology , McGill University , Montreal , Quebec , Canada .
| |
Collapse
|
12
|
Feord HK, Dear FEG, Obbard DJ, van Ooijen G. A Magnesium Transport Protein Related to Mammalian SLC41 and Bacterial MgtE Contributes to Circadian Timekeeping in a Unicellular Green Alga. Genes (Basel) 2019; 10:genes10020158. [PMID: 30791470 PMCID: PMC6410215 DOI: 10.3390/genes10020158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 01/29/2019] [Accepted: 02/12/2019] [Indexed: 11/16/2022] Open
Abstract
Circadian clocks in eukaryotes involve both transcriptional-translational feedback loops, post-translational regulation, and metabolic, non-transcriptional oscillations. We recently identified the involvement of circadian oscillations in the intracellular concentrations of magnesium ions (Mg2+i) that were conserved in three eukaryotic kingdoms. Mg2+i in turn contributes to transcriptional clock properties of period and amplitude, and can function as a zeitgeber to define phase. However, the mechanism-or mechanisms-responsible for the generation of Mg2+i oscillations, and whether these are functionally conserved across taxonomic groups, remain elusive. We employed the cellular clock model Ostreococcustauri to provide a first study of an MgtE domain-containing protein in the green lineage. OtMgtE shares homology with the mammalian SLC41A1 magnesium/sodium antiporter, which has previously been implicated in maintaining clock period. Using genetic overexpression, we found that OtMgtE contributes to both timekeeping and daily changes in Mg2+i. However, pharmacological experiments and protein sequence analyses indicated that critical differences exist between OtMgtE and either the ancestral MgtE channel or the mammalian SLC41 antiporters. We concluded that even though MgtE domain-containing proteins are only distantly related, these proteins retain a shared role in contributing to cellular timekeeping and the regulation of Mg2+i.
Collapse
Affiliation(s)
- Helen K Feord
- School of Biological Sciences, University of Edinburgh, Edinburgh, EH9 3BF, UK.
| | - Frederick E G Dear
- School of Biological Sciences, University of Edinburgh, Edinburgh, EH9 3BF, UK.
| | - Darren J Obbard
- School of Biological Sciences, University of Edinburgh, Edinburgh, EH9 3BF, UK.
| | - Gerben van Ooijen
- School of Biological Sciences, University of Edinburgh, Edinburgh, EH9 3BF, UK.
| |
Collapse
|
13
|
Magnesium-sensitive upstream ORF controls PRL phosphatase expression to mediate energy metabolism. Proc Natl Acad Sci U S A 2019; 116:2925-2934. [PMID: 30718434 DOI: 10.1073/pnas.1815361116] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Phosphatases of regenerating liver (PRL-1, PRL-2, and PRL-3, also known as PTP4A1, PTP4A2, and PTP4A3) control magnesium homeostasis through an association with the CNNM magnesium transport regulators. Although high PRL levels have been linked to cancer progression, regulation of their expression is poorly understood. Here we show that modulating intracellular magnesium levels correlates with a rapid change of PRL expression by a mechanism involving its 5'UTR mRNA region. Mutations or CRISPR-Cas9 targeting of the conserved upstream ORF present in the mRNA leader derepress PRL protein synthesis and attenuate the translational response to magnesium levels. Mechanistically, magnesium depletion reduces intracellular ATP but up-regulates PRL protein expression via activation of the AMPK/mTORC2 pathway, which controls cellular energy status. Hence, altered PRL-2 expression leads to metabolic reprogramming of the cells. These findings uncover a magnesium-sensitive mechanism controlling PRL expression, which plays a role in cellular bioenergetics.
Collapse
|
14
|
Hardy S, Kostantin E, Hatzihristidis T, Zolotarov Y, Uetani N, Tremblay ML. Physiological and oncogenic roles of thePRLphosphatases. FEBS J 2018; 285:3886-3908. [DOI: 10.1111/febs.14503] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/30/2018] [Accepted: 05/09/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Serge Hardy
- Rosalind and Morris Goodman Cancer Research Centre Montréal Canada
| | - Elie Kostantin
- Rosalind and Morris Goodman Cancer Research Centre Montréal Canada
- Department of Biochemistry McGill University Montréal Canada
| | - Teri Hatzihristidis
- Rosalind and Morris Goodman Cancer Research Centre Montréal Canada
- Department of Medicine Division of Experimental Medicine McGill University Montreal Canada
| | - Yevgen Zolotarov
- Rosalind and Morris Goodman Cancer Research Centre Montréal Canada
- Department of Biochemistry McGill University Montréal Canada
| | - Noriko Uetani
- Rosalind and Morris Goodman Cancer Research Centre Montréal Canada
| | - Michel L. Tremblay
- Rosalind and Morris Goodman Cancer Research Centre Montréal Canada
- Department of Biochemistry McGill University Montréal Canada
- Department of Medicine Division of Experimental Medicine McGill University Montreal Canada
| |
Collapse
|
15
|
Silver R. Cells have sex chromosomes and circadian clocks: Implications for organismal level functions. Physiol Behav 2018; 187:6-12. [DOI: 10.1016/j.physbeh.2017.11.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/24/2017] [Accepted: 11/14/2017] [Indexed: 10/18/2022]
|
16
|
Maguire D, Neytchev O, Talwar D, McMillan D, Shiels PG. Telomere Homeostasis: Interplay with Magnesium. Int J Mol Sci 2018; 19:E157. [PMID: 29303978 PMCID: PMC5796106 DOI: 10.3390/ijms19010157] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 12/21/2017] [Accepted: 01/03/2018] [Indexed: 12/14/2022] Open
Abstract
Telomere biology, a key component of the hallmarks of ageing, offers insight into dysregulation of normative ageing processes that accompany age-related diseases such as cancer. Telomere homeostasis is tightly linked to cellular metabolism, and in particular with mitochondrial physiology, which is also diminished during cellular senescence and normative physiological ageing. Inherent in the biochemistry of these processes is the role of magnesium, one of the main cellular ions and an essential cofactor in all reactions that use ATP. Magnesium plays an important role in many of the processes involved in regulating telomere structure, integrity and function. This review explores the mechanisms that maintain telomere structure and function, their influence on circadian rhythms and their impact on health and age-related disease. The pervasive role of magnesium in telomere homeostasis is also highlighted.
Collapse
Affiliation(s)
- Donogh Maguire
- Emergency Medicine Department, Glasgow Royal Infirmary, Glasgow G4 0SF, UK.
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow G4 0SF, UK.
| | - Ognian Neytchev
- Section of Epigenetics, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK.
| | - Dinesh Talwar
- The Scottish Trace Element and Micronutrient Reference Laboratory, Department of Biochemistry, Royal Infirmary, Glasgow G31 2ER, UK.
| | - Donald McMillan
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow G4 0SF, UK.
| | - Paul G Shiels
- Section of Epigenetics, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK.
| |
Collapse
|
17
|
Mining the Complex Family of Protein Tyrosine Phosphatases for Checkpoint Regulators in Immunity. Curr Top Microbiol Immunol 2017; 410:191-214. [PMID: 28929190 DOI: 10.1007/82_2017_68] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The family of protein tyrosine phosphatases (PTPs) includes 107 genes in humans that are diverse in their structures and expression profiles. The majority are present in immune cells and play various roles in either inhibiting or promoting the duration and amplitude of signaling cascades. Several PTPs, including TC-PTP (PTPN2) and SHP-1 (PTPN6), have been recognized as being crucial for maintaining proper immune response and self-tolerance, and have gained recognition as true immune system checkpoint modulators. This chapter details the most recent literature on PTPs and immunity by examining their known functions in regulating signaling from either established checkpoint inhibitors or by their intrinsic properties, as modulators of the immune response. Notably, we review PTP regulatory properties in macrophages, antigen-presenting dendritic cells, and T cells. Overall, we present the PTP gene family as a remarkable source of novel checkpoint inhibitors wherein lies a great number of new targets for immunotherapies.
Collapse
|