1
|
Anderson CM, Kulkarni A, Maier B, Huang F, Figatner K, Chakraborty A, Pratuangtham S, May SC, Tersey SA, Anderson RM, Mirmira RG. Hypusinated and unhypusinated isoforms of the translation factor eIF5A exert distinct effects in models of pancreas development and function. J Biol Chem 2025; 301:108209. [PMID: 39832654 PMCID: PMC11869520 DOI: 10.1016/j.jbc.2025.108209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/30/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Hypusination of eukaryotic translation initiation factor 5A (eIF5A) is essential for its role in translation elongation and termination. Although the function of hypusinated eIF5A (eIF5AHyp) in cellular proliferation is well characterized, the role of its unhypusinated form (eIF5ALys) remains unclear. We hypothesized that eIF5ALys exerts independent and negative effects on cellular replication and metabolism, distinct from the loss of eIF5AHyp. To test this hypothesis, we utilized zebrafish and mouse models with inducible knockdowns of deoxyhypusine synthase (DHPS) and eIF5A to investigate their roles in cellular growth. Gene expression analysis via RNA sequencing and morphometric measurements of pancreas and β-cell mass were performed to assess phenotypic changes and identify affected biological pathways. Loss of DHPS in zebrafish resulted in significant defects in pancreatic growth, accompanied by changes in gene expression related to mRNA translation, neurogenesis, and stress pathways. By contrast, knockdown of eIF5A had minimal impact on pancreas development, suggesting that the effects of DHPS loss are not solely because of the lack of eIF5AHyp. In mice, β-cell-specific deletion of DHPS impaired β-cell mass expansion and glucose tolerance, whereas eIF5A deletion had no statistically significant effects. These findings provide evidence for an independent role for eIF5ALys in regulating developmental and functional responses in pancreas health and disease.
Collapse
Affiliation(s)
- Cara M Anderson
- Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA; Biological Sciences Division, The University of Chicago, Chicago, Illinois, USA
| | - Abhishek Kulkarni
- Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Bernhard Maier
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Fei Huang
- Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Kayla Figatner
- Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | | | | | - Sarah C May
- Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA
| | - Sarah A Tersey
- Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA; Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Ryan M Anderson
- Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA; Department of Medicine, The University of Chicago, Chicago, Illinois, USA.
| | - Raghavendra G Mirmira
- Kovler Diabetes Center, The University of Chicago, Chicago, Illinois, USA; Biological Sciences Division, The University of Chicago, Chicago, Illinois, USA; Department of Medicine, The University of Chicago, Chicago, Illinois, USA; Department of Pediatrics, The University of Chicago, Chicago, Illinois, USA.
| |
Collapse
|
2
|
Zhang X, Qian M, Liu M, He M, Li FR, Zheng L. The Associations of Dietary Polyamines with Incident Type 2 Diabetes Mellitus: A Large Prospective Cohort Study. Nutrients 2025; 17:186. [PMID: 39796620 PMCID: PMC11722915 DOI: 10.3390/nu17010186] [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: 11/29/2024] [Revised: 12/29/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
OBJECTIVES This study aimed to analyze the associations between dietary polyamine intake and incident T2DM. METHODS This prospective analysis included 168,137 participants from the UK Biobank who did not have T2DM at baseline. Dietary polyamines were calculated based on portion sizes of food items and a nutrient database. Incident T2DM was defined by hospital admissions with ICD10 codes E11-E14. Cox proportional hazard regression models and restricted cubic splines were used to examine the associations between dietary polyamine intake and incident T2DM. RESULTS During a median follow-up of 11.2 years (IQR, 11.8-13.2), 4422 (2.6%) participants developed T2DM. The average (SD) daily dietary intake was 10.5 (11.8) mg/day for spermidine, 4.3 (2.1) mg/day for spermine, and 12.7 (6.9) mg/day for putrescine. Compared to quintile 1, the multivariable-adjusted hazard ratios (95% CI) for quintiles 2-5 of dietary spermidine were 0.87 (0.79 to 0.96), 0.87 (0.79 to 0.96), 0.91 (0.82 to 0.99), and 0.96 (0.88 to 1.06); for dietary spermine, they were 1.01 (0.91 to 1.11), 1.03 (0.93 to 1.13), 1.07 (0.97 to 1.18), and 1.11 (1.01 to 1.23); and for dietary putrescine, they were 0.84 (0.76 to 0.92), 0.83 (0.79 to 0.91), 0.82 (0.74 to 0.90), and 0.87 (0.80 to 0.96). CONCLUSIONS Higher dietary spermidine and putrescine were associated with a lower risk of T2DM, while higher dietary spermine appeared to be associated with a higher risk of T2DM. These findings suggest optimal levels of dietary polyamine intake and indicate that polyamines may be promising targets for nutritional interventions in the prevention and management of T2DM.
Collapse
Affiliation(s)
- Xiaohong Zhang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (X.Z.); (M.Q.)
| | - Mingxia Qian
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (X.Z.); (M.Q.)
| | - Min Liu
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang 110122, China; (M.L.); (M.H.)
| | - Mengyao He
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang 110122, China; (M.L.); (M.H.)
| | - Fu-Rong Li
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen 518055, China
| | - Liqiang Zheng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (X.Z.); (M.Q.)
| |
Collapse
|
3
|
Zhang T, Fu W, Zhang H, Li J, Xing B, Cai Y, Zhang M, Liu X, Qi C, Qian L, Hu X, Zhu H, Yang S, Zhang M, Liu J, Li G, Li Y, Xiang R, Qi Z, Hu J, Li Y, Zou C, Wang Q, Jin X, Pang R, Li P, Liu J, Zhang Y, Wang Z, Zhu ZJ, Shan B, Yuan J. Spermidine mediates acetylhypusination of RIPK1 to suppress diabetes onset and progression. Nat Cell Biol 2024; 26:2099-2114. [PMID: 39511379 DOI: 10.1038/s41556-024-01540-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 09/19/2024] [Indexed: 11/15/2024]
Abstract
It has been established that N-acetyltransferase (murine NAT1 (mNAT1) and human NAT2 (hNAT2)) mediates insulin sensitivity in type 2 diabetes. Here we show that mNAT1 deficiency leads to a decrease in cellular spermidine-a natural polyamine exhibiting health-protective and anti-ageing effects-but understanding of its mechanism is limited. We identify that mNAT1 and hNAT2 modulate a type of post-translational modification involving acetylated spermidine, which we name acetylhypusination, on receptor-interacting serine/threonine-protein kinase 1 (RIPK1)-a key regulator of inflammation and cell death. Spermidine supplementation decreases RIPK1-mediated cell death and diabetic phenotypes induced by NAT1 deficiency in vivo. Furthermore, insulin resistance and diabetic kidney disease mediated by vascular pathology in NAT1-deficient mice can be blocked by inhibiting RIPK1. Finally, we demonstrate a decrease in spermidine and activation of RIPK1 in the vascular tissues of human patients with diabetes. Our study suggests a role for vascular pathology in diabetes onset and progression and identifies the inhibition of RIPK1 kinase as a potential therapeutic approach for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Tian Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Weixin Fu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- Nankai University, Tianjin, China
| | - Haosong Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianlong Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Beizi Xing
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuping Cai
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Mengmeng Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Xuheng Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chunting Qi
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Lihui Qian
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Xinbo Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Hua Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Shuailong Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Min Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianping Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Ganquan Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yang Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Rong Xiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zhengqiang Qi
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Junhao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Ying Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Chengyu Zou
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- Shanghai Key Laboratory of Aging Studies, Shanghai, China
| | - Qin Wang
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Shanghai Peritoneal Dialysis Research Center, Renji Hospital, Uremia Diagnosis and Treatment Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Jin
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Pang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junli Liu
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaoyang Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- Shanghai Key Laboratory of Aging Studies, Shanghai, China
| | - Zhaoyin Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zheng-Jiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- Shanghai Key Laboratory of Aging Studies, Shanghai, China.
| | - Bing Shan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| | - Junying Yuan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- Shanghai Key Laboratory of Aging Studies, Shanghai, China.
| |
Collapse
|
4
|
Mi J, Ren L, Andersson O. Leveraging zebrafish to investigate pancreatic development, regeneration, and diabetes. Trends Mol Med 2024; 30:932-949. [PMID: 38825440 DOI: 10.1016/j.molmed.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 06/04/2024]
Abstract
The zebrafish has become an outstanding model for studying organ development and tissue regeneration, which is prominently leveraged for studies of pancreatic development, insulin-producing β-cells, and diabetes. Although studied for more than two decades, many aspects remain elusive and it has only recently been possible to investigate these due to technical advances in transcriptomics, chemical-genetics, genome editing, drug screening, and in vivo imaging. Here, we review recent findings on zebrafish pancreas development, β-cell regeneration, and how zebrafish can be used to provide novel insights into gene functions, disease mechanisms, and therapeutic targets in diabetes, inspiring further use of zebrafish for the development of novel therapies for diabetes.
Collapse
Affiliation(s)
- Jiarui Mi
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden; Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, China.
| | - Lipeng Ren
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden; Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden; Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden.
| |
Collapse
|
5
|
Shojaeinia E, Mastracci TL, Soliman R, Devinsky O, Esguerra CV, Crawford AD. Deoxyhypusine synthase deficiency syndrome zebrafish model: aberrant morphology, epileptiform activity, and reduced arborization of inhibitory interneurons. Mol Brain 2024; 17:68. [PMID: 39334388 PMCID: PMC11429087 DOI: 10.1186/s13041-024-01139-w] [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: 05/27/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
DHPS deficiency syndrome is an ultra-rare neurodevelopmental disorder (NDD) which results from biallelic mutations in the gene encoding the enzyme deoxyhypusine synthase (DHPS). DHPS is essential to synthesize hypusine, a rare amino acid formed by post-translational modification of a conserved lysine in eukaryotic initiation factor 5 A (eIF5A). DHPS deficiency syndrome causes epilepsy, cognitive and motor impairments, and mild facial dysmorphology. In mice, a brain-specific genetic deletion of Dhps at birth impairs eIF5AHYP-dependent mRNA translation. This alters expression of proteins required for neuronal development and function, and phenotypically models features of human DHPS deficiency. We studied the role of DHPS in early brain development using a zebrafish loss-of-function model generated by knockdown of dhps expression with an antisense morpholino oligomer (MO) targeting the exon 2/intron 2 (E2I2) splice site of the dhps pre-mRNA. dhps knockdown embryos exhibited dose-dependent developmental delay and dysmorphology, including microcephaly, axis truncation, and body curvature. In dhps knockdown larvae, electrophysiological analysis showed increased epileptiform activity, and confocal microscopy analysis revealed reduced arborisation of GABAergic neurons. Our findings confirm that hypusination of eIF5A by DHPS is needed for early brain development, and zebrafish with an antisense knockdown of dhps model features of DHPS deficiency syndrome.
Collapse
Affiliation(s)
- Elham Shojaeinia
- Center for Molecular Medicine Norway (NCMM), University of Oslo, Oslo, Norway
- Institute for Orphan Drug Discovery, Bremerhaven, Germany
| | - Teresa L Mastracci
- Department of Biology, Indiana University-Indianapolis, Indianapolis, IN, USA
| | - Remon Soliman
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Orrin Devinsky
- Department of Neurology, New York University Langone Medical Center, New York, NY, USA
| | - Camila V Esguerra
- Center for Molecular Medicine Norway (NCMM), University of Oslo, Oslo, Norway
| | - Alexander D Crawford
- Institute for Orphan Drug Discovery, Bremerhaven, Germany.
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg.
| |
Collapse
|
6
|
Connors CT, Villaca CB, Anderson-Baucum EK, Rosario SR, Rutan CD, Childress PJ, Padgett LR, Robertson MA, Mastracci TL. A Translational Regulatory Mechanism Mediated by Hypusinated Eukaryotic Initiation Factor 5A Facilitates β-Cell Identity and Function. Diabetes 2024; 73:461-473. [PMID: 38055903 PMCID: PMC10882153 DOI: 10.2337/db23-0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
As professional secretory cells, β-cells require adaptable mRNA translation to facilitate a rapid synthesis of proteins, including insulin, in response to changing metabolic cues. Specialized mRNA translation programs are essential drivers of cellular development and differentiation. However, in the pancreatic β-cell, the majority of factors identified to promote growth and development function primarily at the level of transcription. Therefore, despite its importance, the regulatory role of mRNA translation in the formation and maintenance of functional β-cells is not well defined. In this study, we have identified a translational regulatory mechanism mediated by the specialized mRNA translation factor eukaryotic initiation factor 5A (eIF5A), which facilitates the maintenance of β-cell identity and function. The mRNA translation function of eIF5A is only active when it is posttranslationally modified ("hypusinated") by the enzyme deoxyhypusine synthase (DHPS). We have discovered that the absence of β-cell DHPS in mice reduces the synthesis of proteins critical to β-cell identity and function at the stage of β-cell maturation, leading to a rapid and reproducible onset of diabetes. Therefore, our work has revealed a gatekeeper of specialized mRNA translation that permits the β-cell, a metabolically responsive secretory cell, to maintain the integrity of protein synthesis necessary during times of induced or increased demand. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Craig T. Connors
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN
| | | | | | - Spencer R. Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Caleb D. Rutan
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN
| | | | | | | | - Teresa L. Mastracci
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
7
|
Padgett LR, Shinkle MR, Rosario S, Stewart TM, Foley JR, Casero RA, Park MH, Chung WK, Mastracci TL. Deoxyhypusine synthase mutations alter the post-translational modification of eukaryotic initiation factor 5A resulting in impaired human and mouse neural homeostasis. HGG ADVANCES 2023; 4:100206. [PMID: 37333770 PMCID: PMC10275725 DOI: 10.1016/j.xhgg.2023.100206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
DHPS deficiency is a rare genetic disease caused by biallelic hypomorphic variants in the Deoxyhypusine synthase (DHPS) gene. The DHPS enzyme functions in mRNA translation by catalyzing the post-translational modification, and therefore activation, of eukaryotic initiation factor 5A (eIF5A). The observed clinical outcomes associated with human mutations in DHPS include developmental delay, intellectual disability, and seizures. Therefore, to increase our understanding of this rare disease, it is critical to determine the mechanisms by which mutations in DHPS alter neurodevelopment. In this study, we have generated patient-derived lymphoblast cell lines and demonstrated that human DHPS variants alter DHPS protein abundance and impair enzyme function. Moreover, we observe a shift in the abundance of the post-translationally modified forms of eIF5A; specifically, an increase in the nuclear localized acetylated form (eIF5AAcK47) and concomitant decrease in the cytoplasmic localized hypusinated form (eIF5AHYP). Generation and characterization of a mouse model with a genetic deletion of Dhps in the brain at birth shows that loss of hypusine biosynthesis impacts neuronal function due to impaired eIF5AHYP-dependent mRNA translation; this translation defect results in altered expression of proteins required for proper neuronal development and function. This study reveals new insight into the biological consequences and molecular impact of human DHPS deficiency and provides valuable information toward the goal of developing treatment strategies for this rare disease.
Collapse
Affiliation(s)
- Leah R. Padgett
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| | - Mollie R. Shinkle
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Spencer Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Tracy Murray Stewart
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jackson R. Foley
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Robert A. Casero
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Myung Hee Park
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4340, USA
| | - Wendy K. Chung
- Departments of Pediatrics and Medicine, Columbia University, New York, NY 10032, USA
| | - Teresa L. Mastracci
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
8
|
Connors CT, Anderson-Baucum EK, Rosario S, Villaca CBP, Rutan CD, Childress PJ, Padgett LR, Robertson MA, Mastracci TL. Deoxyhypusine synthase is required for the translational regulation of pancreatic beta cell maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.24.537996. [PMID: 37162889 PMCID: PMC10168283 DOI: 10.1101/2023.04.24.537996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
As professional secretory cells, beta cells require adaptable mRNA translation to facilitate a rapid synthesis of proteins, including insulin, in response to changing metabolic cues. Specialized mRNA translation programs are essential drivers of cellular development and differentiation. However, in the pancreatic beta cell, the majority of factors identified to promote growth and development function primarily at the level of transcription. Therefore, despite its importance, the regulatory role of mRNA translation in the formation and maintenance of functional beta cells is not well defined. In this study, we have identified a translational regulatory mechanism in the beta cell driven by the specialized mRNA translation factor, eukaryotic initiation factor 5A (eIF5A), which facilitates beta cell maturation. The mRNA translation function of eIF5A is only active when it is post-translationally modified ("hypusinated") by the enzyme deoxyhypusine synthase (DHPS). We have discovered that the absence of beta cell DHPS in mice reduces the synthesis of proteins critical to beta cell identity and function at the stage of beta cell maturation, leading to a rapid and reproducible onset of diabetes. Therefore, our work has revealed a gatekeeper of specialized mRNA translation that permits the beta cell, a metabolically responsive secretory cell, to maintain the integrity of protein synthesis necessary during times of induced or increased demand. ARTICLE HIGHLIGHTS Pancreatic beta cells are professional secretory cells that require adaptable mRNA translation for the rapid, inducible synthesis of proteins, including insulin, in response to changing metabolic cues. Our previous work in the exocrine pancreas showed that development and function of the acinar cells, which are also professional secretory cells, is regulated at the level of mRNA translation by a specialized mRNA translation factor, eIF5A HYP . We hypothesized that this translational regulation, which can be a response to stress such as changes in growth or metabolism, may also occur in beta cells. Given that the mRNA translation function of eIF5A is only active when the factor is post-translationally modified ("hypusinated") by the enzyme deoxyhypusine synthase (DHPS), we asked the question: does DHPS/eIF5A HYP regulate the formation and maintenance of functional beta cells? We discovered that in the absence of beta cell DHPS in mice, eIF5A is not hypusinated (activated), which leads to a reduction in the synthesis of critical beta cell proteins that interrupts pathways critical for identity and function. This translational regulation occurs at weaning age, which is a stage of cellular stress and maturation for the beta cell. Therefore without DHPS/eIF5A HYP , beta cells do not mature and mice progress to hyperglycemia and diabetes. Our findings suggest that secretory cells have a mechanism to regulate mRNA translation during times of cellular stress. Our work also implies that driving an increase in mRNA translation in the beta cell might overcome or possibly reverse the beta cell defects that contribute to early dysfunction and the progression to diabetes.
Collapse
|
9
|
Schultz CR, Sheldon RD, Xie H, Demireva EY, Uhl KL, Agnew DW, Geerts D, Bachmann AS. New K50R mutant mouse models reveal impaired hypusination of eif5a2 with alterations in cell metabolite landscape. Biol Open 2023; 12:bio059647. [PMID: 36848144 PMCID: PMC10084858 DOI: 10.1242/bio.059647] [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: 09/21/2022] [Accepted: 02/21/2023] [Indexed: 03/01/2023] Open
Abstract
The eukaryotic translation initiation factor 5A1 (eIF5A1) and 5A2 (eIF5A2) are important proteins in a variety of physiological and pathophysiological processes and their function has been linked to neurodevelopmental disorders, cancer, and viral infections. Here, we report two new genome-edited mouse models, generated using a CRISPR-Cas9 approach, in which the amino acid residue lysine 50 is replaced with arginine 50 (K50R) in eIF5A1 or in the closely related eIF5A2 protein. This mutation prevents the spermidine-dependent post-translational formation of hypusine, a unique lysine derivative that is necessary for activation of eIF5A1 and eIF5A2. Mouse brain lysates from homozygous eif5a2-K50R mutant mice (eif5a2K50R/K50R) confirmed the absence of hypusine formation of eIF5A2, and metabolomic analysis of primary mouse dermal fibroblasts revealed significant alterations in the metabolite landscape compared to controls including increased levels of tryptophan, kyrunenine, pyridoxine, nicotinamide adenine dinucleotide, riboflavin, flavin adenine dinucleotide, pantothenate, and coenzyme A. Further supported by new publicly available bioinformatics data, these new mouse models represent excellent in vivo models to study hypusine-dependent biological processes, hypusination-related disorders caused by eIF5A1 and eIF5A2 gene aberrations or mRNA expression dysregulation, as well as several major human cancer types and potential therapies.
Collapse
Affiliation(s)
- Chad R. Schultz
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Ryan D. Sheldon
- Core Technologies and Services, Mass Spectrometry Core, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Huirong Xie
- Transgenic and Genome Editing Facility, Institute for Quantitative Health Science and Engineering, Research Technology Support Facility, Michigan State University, East Lansing, MI 48824, USA
| | - Elena Y. Demireva
- Transgenic and Genome Editing Facility, Institute for Quantitative Health Science and Engineering, Research Technology Support Facility, Michigan State University, East Lansing, MI 48824, USA
| | - Katie L. Uhl
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Dalen W. Agnew
- Department of Pathobiology & Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Dirk Geerts
- Department of Hematology, Amsterdam University Medical Center, Location VUMC, 1081 HV Amsterdam, The Netherlands
| | - André S. Bachmann
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| |
Collapse
|
10
|
Kulkarni A, Anderson CM, Mirmira RG, Tersey SA. Role of Polyamines and Hypusine in β Cells and Diabetes Pathogenesis. Metabolites 2022; 12:344. [PMID: 35448531 PMCID: PMC9028953 DOI: 10.3390/metabo12040344] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 02/04/2023] Open
Abstract
The polyamines-putrescine, spermidine, and spermine-are polycationic, low molecular weight amines with cellular functions primarily related to mRNA translation and cell proliferation. Polyamines partly exert their effects via the hypusine pathway, wherein the polyamine spermidine provides the aminobutyl moiety to allow posttranslational modification of the translation factor eIF5A with the rare amino acid hypusine (hydroxy putrescine lysine). The "hypusinated" eIF5A (eIF5Ahyp) is considered to be the active form of the translation factor necessary for the translation of mRNAs associated with stress and inflammation. Recently, it has been demonstrated that activity of the polyamines-hypusine circuit in insulin-producing islet β cells contributes to diabetes pathogenesis under conditions of inflammation. Elevated levels of polyamines are reported in both exocrine and endocrine cells of the pancreas, which may contribute to endoplasmic reticulum stress, oxidative stress, inflammatory response, and autophagy. In this review, we have summarized the existing research on polyamine-hypusine metabolism in the context of β-cell function and diabetes pathogenesis.
Collapse
Affiliation(s)
| | | | | | - Sarah A. Tersey
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA; (A.K.); (C.M.A.); (R.G.M.)
| |
Collapse
|
11
|
Rawson A, Saxena V, Gao H, Hooks J, Xuei X, McGuire P, Hato T, Hains DS, Anderson RM, Schwaderer AL. A Pilot Single Cell Analysis of the Zebrafish Embryo Cellular Responses to Uropathogenic Escherichia coli Infection. Pathog Immun 2022; 7:1-18. [PMID: 35178490 PMCID: PMC8843076 DOI: 10.20411/pai.v7i1.479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/06/2021] [Indexed: 11/23/2022] Open
Abstract
Background: Uropathogenic Escherichia coli (UPEC) infections are common and when they disseminate can be of high morbidity.
Methods: We studied the effects of UPEC infection using single cell RNA sequencing (scRNAseq) in zebrafish. Bulk RNA sequencing has historically been used to evaluate gene expression patterns, but scRNAseq allows gene expression to be evaluated at the single cell level and is optimal for evaluating heterogeneity within cell types and rare cell types. Zebrafish cohorts were injected with either saline or UPEC,and scRNAseq and canonical pathway analyses were performed.
Results: Canonical pathway analysis of scRNAseq data provided key information regarding innate immune pathways in the cells determined to be thymus cells, ionocytes, macrophages/monocytes, and pronephros cells. Pathways activated in thymus cells included interleukin 6 (IL-6) signaling and production of reactive oxygen species. Fc receptor-mediated phagocytosis was a leading canonical pathway in the pronephros and macrophages. Genes that were downregulated in UPEC vs saline exposed embryos involved the cellular response to the Gram-negative endotoxin lipopolysaccharide (LPS) and included Forkhead Box O1a (Foxo1a), Tribbles Pseudokinase 3 (Trib3), Arginase 2 (Arg2) and Polo Like Kinase 3 (Plk3).
Conclusions: Because 4-day post fertilization zebrafish embryos only have innate immune systems, the scRNAseq provides insights into pathways and genes that cell types utilize in the bacterial response. Based on our analysis, we have identified genes and pathways that might serve as genetic targets for treatment and further investigation in UPEC infections at the single cell level.
Collapse
Affiliation(s)
- Ashley Rawson
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology
| | - Vijay Saxena
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology
| | - Hongyu Gao
- Indiana University School of Medicine, Department of Medical & Molecular Genetics
| | - Jenaya Hooks
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology
| | - Xiaoling Xuei
- Indiana University School of Medicine, Department of Medical & Molecular Genetics
| | - Patrick McGuire
- Indiana University School of Medicine, Department of Medical & Molecular Genetics
| | - Takashi Hato
- Indiana University School of Medicine, Department of Medicine, Division of Nephrology
| | - David S. Hains
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology
| | - Ryan M. Anderson
- University of Chicago, Section of Endocrinology, Diabetes and Metabolism
- CORRESPONDING AUTHOR Andrew Schwaderer, Indiana University School of Medicine, Riley Hospital for Children, 699 Riley Hospital Dr., RR230, Indianapolis, IN 46202; Phone: 317-274-2527;
| | - Andrew L. Schwaderer
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology
- CORRESPONDING AUTHOR Ryan M Anderson, University of Chicago, Medicine-Endocrinology, Chicago, IL 60637;
| |
Collapse
|
12
|
Fahrmann JF, Wasylishen AR, Pieterman CRC, Irajizad E, Vykoukal J, Murage E, Wu R, Dennison JB, Krishna H, Peterson CB, Lozano G, Zhao H, Do KA, Halperin DM, Agarwal SK, Blau JE, Del Rivero J, Nilubol N, Walter MF, Welch JM, Weinstein LS, Vriens MR, van Leeuwaarde RS, van Treijen MJC, Valk GD, Perrier ND, Hanash SM. A Blood-based Polyamine Signature Associated With MEN1 Duodenopancreatic Neuroendocrine Tumor Progression. J Clin Endocrinol Metab 2021; 106:e4969-e4980. [PMID: 34318891 PMCID: PMC8864750 DOI: 10.1210/clinem/dgab554] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Duodenopancreatic neuroendocrine tumors (dpNETs) frequently occur in patients with multiple endocrine neoplasia type 1 (MEN1), and metastatic dpNET is the primary cause of disease-related mortality. There is a need for biomarkers that can identify patients with MEN1-related dpNETs that are at high risk of developing distant metastasis. Polyamines have tumor-promoting roles in several cancer types. OBJECTIVE We hypothesized that MEN1-dpNET-related disease progression is associated with elevated levels of circulating polyamines. METHODS Through an international collaboration between The University of Texas MD Anderson Cancer Center, the National Institutes of Health, and the University Medical Center Utrecht, plasma polyamine levels were assessed using mass spectrometry in 84 patients with MEN1 (20 with distant metastatic dpNETs [patients] and 64 with either indolent dpNETs or no dpNETs [controls]). A mouse model of MEN1-pNET, Men1fl/flPdx1-CreTg, was used to test time-dependent changes in plasma polyamines associated with disease progression. RESULTS A 3-marker plasma polyamine signature (3MP: N-acetylputrescine, acetylspermidine, and diacetylspermidine) distinguished patients with metastatic dpNETs from controls in an initial set of plasmas from the 3 participating centers. The fixed 3MP yielded an area under the curve of 0.84 (95% CI, 0.62-1.00) with 66.7% sensitivity at 95% specificity for distinguishing patients from controls in an independent test set from MDACC. In Men1fl/flPdx1-CreTg mice, the 3MP was elevated early and remained high during disease progression. CONCLUSION Our findings provide a basis for prospective testing of blood-based polyamines as a potential means for monitoring patients with MEN1 for harboring or developing aggressive disease.
Collapse
Affiliation(s)
- Johannes F Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Amanda R Wasylishen
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Carolina R C Pieterman
- Department of Surgical Oncology, Section of Surgical Endocrinology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ehsan Irajizad
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jody Vykoukal
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Eunice Murage
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ranran Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer B Dennison
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hansini Krishna
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Christine B Peterson
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Guillermina Lozano
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hua Zhao
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Family Medicine and Population Health, Virginia Commonwealth University, Richmond, Virgina, USA
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel M Halperin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sunita K Agarwal
- Metabolic Diseases Branch, the National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jenny E Blau
- Metabolic Diseases Branch, the National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jaydira Del Rivero
- Developmental Therapeutics Branch, the National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Naris Nilubol
- Surgical Oncology Program, the National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mary F Walter
- Core for Clinical Laboratory Services, the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - James M Welch
- Metabolic Diseases Branch, the National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Lee S Weinstein
- Metabolic Diseases Branch, the National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Menno R Vriens
- Department of Surgical Oncology and Endocrine Surgery, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
- Center for Neuroendocrine Tumors, ENETS Center of Excellence, Netherlands Cancer Institute Amsterdam, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Rachel S van Leeuwaarde
- Center for Neuroendocrine Tumors, ENETS Center of Excellence, Netherlands Cancer Institute Amsterdam, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
- Department of Endocrine Oncology, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Mark J C van Treijen
- Center for Neuroendocrine Tumors, ENETS Center of Excellence, Netherlands Cancer Institute Amsterdam, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
- Department of Endocrine Oncology, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Gerlof D Valk
- Center for Neuroendocrine Tumors, ENETS Center of Excellence, Netherlands Cancer Institute Amsterdam, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
- Department of Endocrine Oncology, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Nancy D Perrier
- Department of Surgical Oncology, Section of Surgical Endocrinology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Correspondence: Samir M. Hanash, MD, PhD, Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, 6767 Bertner Ave, Houston, TX 77030, USA.
| |
Collapse
|
13
|
Chang CY, Park JH, Ouh IO, Gu NY, Jeong SY, Lee SA, Lee YH, Hyun BH, Kim KS, Lee J. Novel method to repair articular cartilage by direct reprograming of prechondrogenic mesenchymal stem cells. Eur J Pharmacol 2021; 911:174416. [PMID: 34606836 DOI: 10.1016/j.ejphar.2021.174416] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 12/27/2022]
Abstract
Age-related cartilage loss is worsened by the limited regenerative capacity of chondrocytes. The role of cell-based therapies using mesenchymal stem cells is gaining interest. Adipose tissue-derived mesenchymal stem cells (ADSCs) are an attractive source to generate the optimal number of chondrocytes required to repair a cartilage defect and regenerate hyaline articular cartilage. Here, we report an outstanding technique to prepare chondrocytes for cartilage repair using canine ADSCs. We hypothesized that external electrical fields promote prechondrogenic condensation without requiring genetic modifications or exogenous factors. We analyzed the effect of electrical stimulation (ES) on the differentiation of ADSC micromass into chondrocytes. Highly compact structures were formed within 3 days of ES of canine ADSC micromass. The expression of type I collagen gene was abolished in these cells compared with that in control micromass cultures and monolayer cultures. We further found that ES enhanced the production of proteoglycan, a highly produced extracellular matrix component in chondrocytes. Additionally, single-cell RNA sequencing analysis showed that canine ADSC micromass undergoing ES developed a prechondrogenic cell aggregation, suggesting their metabolic conversion, biogenesis, and calcium ion change. Collectively, our findings demonstrate the capacity of ES to drive the chondrogenesis of ADSCs in the absence of exogenous factors and confirm its commercial potential as a budget-friendly therapy for the repair of cartilage defects.
Collapse
Affiliation(s)
- Chi Young Chang
- Hanyang Digitech, 332-7, Samsung 1-ro, Hwaseong, Gyeonggi-do, 18380, Republic of Korea; Youth Bio Global, 273, Digital-ro, Guro-gu, Seoul, 08381, Republic of Korea
| | - Ju Hyun Park
- Hanyang Digitech, 332-7, Samsung 1-ro, Hwaseong, Gyeonggi-do, 18380, Republic of Korea; Youth Bio Global, 273, Digital-ro, Guro-gu, Seoul, 08381, Republic of Korea
| | - In-Ohk Ouh
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Na-Yeon Gu
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea
| | - So Yeon Jeong
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Se-A Lee
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Yoon-Hee Lee
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Bang-Hun Hyun
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Ki Suk Kim
- Hanyang Digitech, 332-7, Samsung 1-ro, Hwaseong, Gyeonggi-do, 18380, Republic of Korea
| | - Jienny Lee
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon, Gyeongsangbuk-do, 39660, Republic of Korea; Division of Regenerative Medicine Safety Control, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, 187 Osongsaengmyeong 2-ro, Cheongju, Chungcheongbuk-do, 28159, Republic of Korea.
| |
Collapse
|
14
|
Kar RK, Hanner AS, Starost MF, Springer D, Mastracci TL, Mirmira RG, Park MH. Neuron-specific ablation of eIF5A or deoxyhypusine synthase leads to impairments in growth, viability, neurodevelopment, and cognitive functions in mice. J Biol Chem 2021; 297:101333. [PMID: 34688659 PMCID: PMC8605248 DOI: 10.1016/j.jbc.2021.101333] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/13/2021] [Accepted: 10/19/2021] [Indexed: 12/05/2022] Open
Abstract
Eukaryotic initiation factor 5A (eIF5A)†,‡ is an essential protein that requires a unique amino acid, hypusine, for its activity. Hypusine is formed exclusively in eIF5A post-translationally via two enzymes, deoxyhypusine synthase (DHPS) and deoxyhypusine hydroxylase. Each of the genes encoding these proteins, Eif5a, Dhps, and Dohh, is required for mouse embryonic development. Variants in EIF5A or DHPS were recently identified as the genetic basis underlying certain rare neurodevelopmental disorders in humans. To investigate the roles of eIF5A and DHPS in brain development, we generated four conditional KO mouse strains using the Emx1-Cre or Camk2a-Cre strains and examined the effects of temporal- and region-specific deletion of Eif5a or Dhps. The conditional deletion of Dhps or Eif5a by Emx1 promotor-driven Cre expression (E9.5, in the cortex and hippocampus) led to gross defects in forebrain development, reduced growth, and premature death. On the other hand, the conditional deletion of Dhps or Eif5a by Camk2a promoter-driven Cre expression (postnatal, mainly in the CA1 region of the hippocampus) did not lead to global developmental defects; rather, these KO animals exhibited severe impairment in spatial learning, contextual learning, and memory when subjected to the Morris water maze and a contextual learning test. In both models, the Dhps-KO mice displayed more severe impairment than their Eif5a-KO counterparts. The observed defects in the brain, global development, or cognitive functions most likely result from translation errors due to a deficiency in active, hypusinated eIF5A. Our study underscores the important roles of eIF5A and DHPS in neurodevelopment.
Collapse
Affiliation(s)
- Rajesh Kumar Kar
- Molecular and Cellular Biochemistry Section, NIDCR, National Institutes of Health, Bethesda, Maryland, USA
| | - Ashleigh S Hanner
- Molecular and Cellular Biochemistry Section, NIDCR, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthew F Starost
- Division of Veterinary Resources, Diagnostic and Research Services Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Danielle Springer
- NHLBI Murine Phenotyping Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Teresa L Mastracci
- Department of Biology, Indiana University-Purdue University-Indianapolis, Indianapolis, Indiana, USA
| | | | - Myung Hee Park
- Molecular and Cellular Biochemistry Section, NIDCR, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
15
|
Anderson-Baucum E, Piñeros AR, Kulkarni A, Webb-Robertson BJ, Maier B, Anderson RM, Wu W, Tersey SA, Mastracci TL, Casimiro I, Scheuner D, Metz TO, Nakayasu ES, Evans-Molina C, Mirmira RG. Deoxyhypusine synthase promotes a pro-inflammatory macrophage phenotype. Cell Metab 2021; 33:1883-1893.e7. [PMID: 34496231 PMCID: PMC8432737 DOI: 10.1016/j.cmet.2021.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/01/2021] [Accepted: 08/05/2021] [Indexed: 12/24/2022]
Abstract
The metabolic inflammation (meta-inflammation) of obesity is characterized by proinflammatory macrophage infiltration into adipose tissue. Catalysis by deoxyhypusine synthase (DHPS) modifies the translation factor eIF5A to generate a hypusine (Hyp) residue. Hypusinated eIF5A (eIF5AHyp) controls the translation of mRNAs involved in inflammation, but its role in meta-inflammation has not been elucidated. Levels of eIF5AHyp were found to be increased in adipose tissue macrophages from obese mice and in murine macrophages activated to a proinflammatory M1-like state. Global proteomics and transcriptomics revealed that DHPS deficiency in macrophages altered the abundance of proteins involved in NF-κB signaling, likely through translational control of their respective mRNAs. DHPS deficiency in myeloid cells of obese mice suppressed M1 macrophage accumulation in adipose tissue and improved glucose tolerance. These findings indicate that DHPS promotes the post-transcriptional regulation of a subset of mRNAs governing inflammation and chemotaxis in macrophages and contributes to a proinflammatory M1-like phenotype.
Collapse
Affiliation(s)
- Emily Anderson-Baucum
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Annie R Piñeros
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Abhishek Kulkarni
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | | | - Bernhard Maier
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ryan M Anderson
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Wenting Wu
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sarah A Tersey
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Teresa L Mastracci
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Isabel Casimiro
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Donalyn Scheuner
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| | - Thomas O Metz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Ernesto S Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Roudebush VA Medical Center, Indianapolis, IN 46202, USA.
| | | |
Collapse
|
16
|
Le Bastard Q, Berthelot L, Soulillou JP, Montassier E. Impact of non-antibiotic drugs on the human intestinal microbiome. Expert Rev Mol Diagn 2021; 21:911-924. [PMID: 34225544 DOI: 10.1080/14737159.2021.1952075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The gut microbiota is composed of trillions of microbial cells and viruses that interact with hosts. The composition of the gut microbiota is influenced by several factors including age, diet, diseases, or medications. The impact of drugs on the microbiota is not limited to antibiotics and many non-antibiotic molecules significantly alter the composition of the intestinal microbiota. AREAS COVERED This review focuses on the impact of four of the most widely prescribed non-antibiotic drugs in the world: Proton-pump inhibitors, metformin, statins, and non-steroidal anti-inflammatory. We conducted a systematic review by searching online databases including Medline, Web of science, and Scopus for indexed articles published in English until February 2021. We included studies assessing the intestinal microbiome alterations associated with proton pump inhibitors (PPIs), metformin, statins, and nonsteroidal anti-inflammatory drugs (NSAIDs). Only studies using culture-independent molecular techniques were included. EXPERT OPINION The taxonomical signature associated with non-antibiotic drugs are not yet fully described, especially in the field of metabolomic. The identification of taxonomic profiles associated a specific molecule provides information on its mechanism of action through interaction with the intestinal microbiota. Many side effects could be related to the dysbiosis induced by these molecules.
Collapse
Affiliation(s)
- Quentin Le Bastard
- Microbiota Hosts Antibiotics and Bacterial Resistances (Mihar), Université De Nantes, Nantes, France.,Service Des Urgences, CHU De Nantes, Nantes, France
| | - Laureline Berthelot
- Centre De Recherche En Transplantation Et Immunologie UMR 1064, INSERM, Université De Nantes, Nantes, France.,Institut De Transplantation Urologie Néphrologie (ITUN), CHU De Nantes, Nantes, France
| | - Jean-Paul Soulillou
- Centre De Recherche En Transplantation Et Immunologie UMR 1064, INSERM, Université De Nantes, Nantes, France.,Institut De Transplantation Urologie Néphrologie (ITUN), CHU De Nantes, Nantes, France
| | - Emmanuel Montassier
- Microbiota Hosts Antibiotics and Bacterial Resistances (Mihar), Université De Nantes, Nantes, France.,Service Des Urgences, CHU De Nantes, Nantes, France
| |
Collapse
|
17
|
Padgett LR, Robertson MA, Anderson‐Baucum EK, Connors CT, Wu W, Mirmira RG, Mastracci TL. Deoxyhypusine synthase, an essential enzyme for hypusine biosynthesis, is required for proper exocrine pancreas development. FASEB J 2021; 35:e21473. [PMID: 33811703 PMCID: PMC8034418 DOI: 10.1096/fj.201903177r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/21/2021] [Accepted: 02/10/2021] [Indexed: 12/17/2022]
Abstract
Pancreatic diseases including diabetes and exocrine insufficiency would benefit from therapies that reverse cellular loss and/or restore cellular mass. The identification of molecular pathways that influence cellular growth is therefore critical for future therapeutic generation. Deoxyhypusine synthase (DHPS) is an enzyme that post-translationally modifies and activates the mRNA translation factor eukaryotic initiation factor 5A (eIF5A). Previous work demonstrated that the inhibition of DHPS impairs zebrafish exocrine pancreas development; however, the link between DHPS, eIF5A, and regulation of pancreatic organogenesis remains unknown. Herein we identified that the conditional deletion of either Dhps or Eif5a in the murine pancreas results in the absence of acinar cells. Because DHPS catalyzes the activation of eIF5A, we evaluated and uncovered a defect in mRNA translation concomitant with defective production of proteins that influence cellular development. Our studies reveal a heretofore unappreciated role for DHPS and eIF5A in the synthesis of proteins required for cellular development and function.
Collapse
Affiliation(s)
| | - Morgan A. Robertson
- Department of BiologyIndiana University‐Purdue University‐Indianapolis (IUPUI)IndianapolisINUSA
| | | | - Craig T. Connors
- Department of BiologyIndiana University‐Purdue University‐Indianapolis (IUPUI)IndianapolisINUSA
| | - Wenting Wu
- Center for Diabetes and Metabolic DiseasesIndiana University School of MedicineIndianapolisINUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisINUSA
| | - Raghavendra G. Mirmira
- Center for Diabetes and Metabolic DiseasesIndiana University School of MedicineIndianapolisINUSA
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisINUSA
- Department of PediatricsIndiana University School of MedicineIndianapolisINUSA
- Kovler Diabetes Center and the Department of MedicineUniversity of ChicagoChicagoILUSA
| | - Teresa L. Mastracci
- Indiana Biosciences Research InstituteIndianapolisINUSA
- Department of BiologyIndiana University‐Purdue University‐Indianapolis (IUPUI)IndianapolisINUSA
- Center for Diabetes and Metabolic DiseasesIndiana University School of MedicineIndianapolisINUSA
- Department of Biochemistry and Molecular BiologyIndiana University School of MedicineIndianapolisINUSA
| |
Collapse
|
18
|
Faundes V, Jennings MD, Crilly S, Legraie S, Withers SE, Cuvertino S, Davies SJ, Douglas AGL, Fry AE, Harrison V, Amiel J, Lehalle D, Newman WG, Newkirk P, Ranells J, Splitt M, Cross LA, Saunders CJ, Sullivan BR, Granadillo JL, Gordon CT, Kasher PR, Pavitt GD, Banka S. Impaired eIF5A function causes a Mendelian disorder that is partially rescued in model systems by spermidine. Nat Commun 2021; 12:833. [PMID: 33547280 PMCID: PMC7864902 DOI: 10.1038/s41467-021-21053-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023] Open
Abstract
The structure of proline prevents it from adopting an optimal position for rapid protein synthesis. Poly-proline-tract (PPT) associated ribosomal stalling is resolved by highly conserved eIF5A, the only protein to contain the amino acid hypusine. We show that de novo heterozygous EIF5A variants cause a disorder characterized by variable combinations of developmental delay, microcephaly, micrognathia and dysmorphism. Yeast growth assays, polysome profiling, total/hypusinated eIF5A levels and PPT-reporters studies reveal that the variants impair eIF5A function, reduce eIF5A-ribosome interactions and impair the synthesis of PPT-containing proteins. Supplementation with 1 mM spermidine partially corrects the yeast growth defects, improves the polysome profiles and restores expression of PPT reporters. In zebrafish, knockdown eif5a partly recapitulates the human phenotype that can be rescued with 1 µM spermidine supplementation. In summary, we uncover the role of eIF5A in human development and disease, demonstrate the mechanistic complexity of EIF5A-related disorder and raise possibilities for its treatment.
Collapse
Affiliation(s)
- Víctor Faundes
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Laboratorio de Genética y Enfermedades Metabólicas, Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
| | - Martin D Jennings
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Siobhan Crilly
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sarah Legraie
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sarah E Withers
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sara Cuvertino
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sally J Davies
- Institute of Medical Genetics, University Hospital of Wales, Cardiff, UK
| | - Andrew G L Douglas
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Andrew E Fry
- Institute of Medical Genetics, University Hospital of Wales, Cardiff, UK
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - Victoria Harrison
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | - Jeanne Amiel
- Department of Genetics, AP-HP, Hôpital Necker Enfants Malades, Paris, France
- 1Laboratory of Embryology and Genetics of Human Malformations, INSERM UMR 1163, Institut Imagine, Paris, France
- Paris Descartes-Sorbonne Paris Cité University, Institut Imagine, Paris, France
| | - Daphné Lehalle
- Department of Genetics, AP-HP, Hôpital Necker Enfants Malades, Paris, France
| | - William G Newman
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK
| | - Patricia Newkirk
- Division of Genetics and Metabolism, Department of Pediatrics, University of South Florida, Tampa, FL, UK
| | - Judith Ranells
- Division of Genetics and Metabolism, Department of Pediatrics, University of South Florida, Tampa, FL, UK
| | - Miranda Splitt
- Northern Genetics Service, Institute of Genetic Medicine, Newcastle upon Tyne, UK
| | - Laura A Cross
- Division of Clinical Genetics, Children's Mercy, Kansas City, MO, USA
- Department of Pediatrics, University of Missour-Kansas City, Kansas City, MO, USA
| | - Carol J Saunders
- Center for Pediatric Genomic Medicine Children's Mercy, Kansas City, MO, USA
- School of Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
- Department of Pathology and Laboratory Medicine, Children's Mercy, Kansas City, MO, USA
| | - Bonnie R Sullivan
- Division of Clinical Genetics, Children's Mercy, Kansas City, MO, USA
- Department of Pediatrics, University of Missour-Kansas City, Kansas City, MO, USA
| | - Jorge L Granadillo
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Christopher T Gordon
- 1Laboratory of Embryology and Genetics of Human Malformations, INSERM UMR 1163, Institut Imagine, Paris, France
- Paris Descartes-Sorbonne Paris Cité University, Institut Imagine, Paris, France
| | - Paul R Kasher
- Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Graham D Pavitt
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
| | - Siddharth Banka
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK.
| |
Collapse
|
19
|
Abstract
Historically, the focus of type II diabetes mellitus (T2DM) research has been on host metabolism and hormone action. However, emerging evidence suggests that the gut microbiome, commensal microbes that colonize the gastrointestinal tract, also play a significant role in T2DM pathogenesis. Specifically, gut microbes metabolize what is available to them through the host diet to produce small molecule metabolites that can have endocrine-like effects on human cells. In fact, the meta-organismal crosstalk between gut microbe-generated metabolites and host receptor systems may represent an untapped therapeutic target for those at risk for or suffering from T2DM. Recent evidence suggests that gut microbe-derived metabolites can impact host adiposity, insulin resistance, and hormone secretion to collectively impact T2DM progression. Here we review the current evidence that structurally diverse gut microbe-derived metabolites, including short chain fatty acids, secondary bile acids, aromatic metabolites, trimethylamine-N-oxide, polyamines, and N-acyl amides, that can engage with host receptors in an endocrine-like manner to promote host metabolic disturbance associated with T2DM. Although these microbe-host signaling circuits are not as well understood as host hormonal signaling, they hold untapped potential as new druggable targets to improve T2DM complications. Whether drugs that selectively target meta-organismal endocrinology will be safe and efficacious in treating T2DM is a key new question in the field of endocrinology. Here we discuss the opportunities and challenges in targeting the gut microbial endocrine organ for the treatment of diabetes and potentially many other diseases where diet-microbe-host interactions play a contributory role.
Collapse
Affiliation(s)
- William Massey
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH, USA
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH, USA
| | - J Mark Brown
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH, USA
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH, USA
- Correspondence: J. Mark Brown, Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
20
|
Hernandez-Perez M, Kulkarni A, Samala N, Sorrell C, El K, Haider I, Aleem AM, Holman TR, Rai G, Tersey SA, Mirmira RG, Anderson RM. A 12-lipoxygenase-Gpr31 signaling axis is required for pancreatic organogenesis in the zebrafish. FASEB J 2020; 34:14850-14862. [PMID: 32918516 PMCID: PMC7606739 DOI: 10.1096/fj.201902308rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 12/19/2022]
Abstract
12-Lipoxygenase (12-LOX) is a key enzyme in arachidonic acid metabolism, and alongside its major product, 12-HETE, plays a key role in promoting inflammatory signaling during diabetes pathogenesis. Although 12-LOX is a proposed therapeutic target to protect pancreatic islets in the setting of diabetes, little is known about the consequences of blocking its enzymatic activity during embryonic development. Here, we have leveraged the strengths of the zebrafish-genetic manipulation and pharmacologic inhibition-to interrogate the role of 12-LOX in pancreatic development. Lipidomics analysis during zebrafish development demonstrated that 12-LOX-generated metabolites of arachidonic acid increase sharply during organogenesis stages, and that this increase is blocked by morpholino-directed depletion of 12-LOX. Furthermore, we found that either depletion or inhibition of 12-LOX impairs both exocrine pancreas growth and unexpectedly, the generation of insulin-producing β cells. We demonstrate that morpholino-mediated knockdown of GPR31, a purported G-protein-coupled receptor for 12-HETE, largely phenocopies both the depletion and the inhibition of 12-LOX. Moreover, we show that loss of GPR31 impairs pancreatic bud fusion and pancreatic duct morphogenesis. Together, these data provide new insight into the requirement of 12-LOX in pancreatic organogenesis and islet formation, and additionally provide evidence that its effects are mediated via a signaling axis that includes the 12-HETE receptor GPR31.
Collapse
Affiliation(s)
- Marimar Hernandez-Perez
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Abhishek Kulkarni
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Niharika Samala
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cody Sorrell
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kimberly El
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Isra Haider
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ansari Mukhtar Aleem
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Theodore R Holman
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Ganesha Rai
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Sarah A Tersey
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Raghavendra G Mirmira
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Medicine, Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Ryan M Anderson
- Department of Pediatrics, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA,Department of Medicine, Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
21
|
Abstract
Type 1 diabetes (T1D) is a disease characterized by destruction of the insulin-producing beta cells. Currently, there remains a critical gap in our understanding of how to reverse or prevent beta cell loss in individuals with T1D. Previous studies in mice discovered that pharmacologically inhibiting polyamine biosynthesis using difluoromethylornithine (DFMO) resulted in preserved beta cell function and mass. Similarly, treatment of non-obese diabetic mice with the tyrosine kinase inhibitor Imatinib mesylate reversed diabetes. The promising findings from these animal studies resulted in the initiation of two separate clinical trials that would repurpose either DFMO (NCT02384889) or Imatinib (NCT01781975) and determine effects on diabetes outcomes; however, whether these drugs directly stimulated beta cell growth remained unknown. To address this, we used the zebrafish model system to determine pharmacological impact on beta cell regeneration. After induction of beta cell death, zebrafish embryos were treated with either DFMO or Imatinib. Neither drug altered whole-body growth or exocrine pancreas length. Embryos treated with Imatinib showed no effect on beta cell regeneration; however, excitingly, DFMO enhanced beta cell regeneration. These data suggest that pharmacological inhibition of polyamine biosynthesis may be a promising therapeutic option to stimulate beta cell regeneration in the setting of diabetes.
Collapse
Affiliation(s)
| | - Leah R. Padgett
- Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Jonathan A. Fine
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Gaurav Chopra
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, USA
- Integrative Data Science Initiative, Purdue University, West Lafayette, IN, USA
| | - Teresa L. Mastracci
- Indiana Biosciences Research Institute, Indianapolis, IN, USA
- Department of Biology, Indiana University, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- CONTACT Teresa L. Mastracci Department of Biology, Indiana University, Indianapolis, IN46202, USA
| |
Collapse
|
22
|
Sato M, Toyama T, Kim MS, Lee JY, Hoshi T, Miura N, Naganuma A, Hwang GW. Increased putrescine levels due to ODC1 overexpression prevents mitochondrial dysfunction-related apoptosis induced by methylmercury. Life Sci 2020; 256:118031. [PMID: 32615186 DOI: 10.1016/j.lfs.2020.118031] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/25/2020] [Accepted: 06/27/2020] [Indexed: 12/23/2022]
Abstract
AIMS We had previously reported that addition of putrescine to the culture medium was reported to reduce methylmercury toxicity in C17.2 neural stem cells. Here, we have examined the inhibition of methylmercury-induced cytotoxicity by putrescine using ODC1-overexpressing C17.2 cells. MATERIALS AND METHODS We established stable ODC1-overexpressing C17.2 cells and evaluated methylmercury-induced apoptosis by examining the TUNEL assay and cleaved caspase-3 levels. Mitochondria-mediated apoptosis was also evaluated by reduction of mitochondrial membrane potential and recruitment of Bax and Bak to the mitochondria. KEY FINDINGS ODC is encoded by ODC1 gene, and putrescine levels in ODC1-overexpressing cells were significantly higher than in control cells. Overexpression of ODC1 and addition of putrescine to the culture medium suppressed DNA fragmentation and caspase-3 activation, which are observed when apoptosis is induced by methylmercury. Moreover, mitochondrial dysfunction and reactive oxygen species (ROS) generation, caused by methylmercury, were also inhibited by the overexpression of ODC1 and putrescine; pretreatment with ODC inhibitor, however, promoted both ROS generation and apoptosis by methylmercury. Finally, we found that Bax and Bak, the apoptosis-promoting factors, to be increased in mitochondria, following methylmercury treatment, and the same was inhibited by overexpression of ODC1. These results suggest that overexpression of ODC1 may prevent mitochondria-mediated apoptosis by methylmercury via increase of putrescine levels. SIGNIFICANCE Our findings provide important clues to clarify mechanisms involved in the defense against methylmercury toxicity and suggest novel biological functions of putrescine.
Collapse
Affiliation(s)
- Masayuki Sato
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Takashi Toyama
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Min-Seok Kim
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; Inhalation Toxicology Research Group, Korea Institute of Toxicology, 30, Baekhak1-gil Jeongeup-si, Jeollabuk-do 56212, Republic of Korea
| | - Jin-Yong Lee
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya 464-8650, Japan
| | - Takayuki Hoshi
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Nobuhiko Miura
- Laboratory of Environmental and Molecular Toxicology, Yokohama University of Pharmacy, 601 Matano-cho, Totsuka-ku, Yokohama, Kanagawa 245-0066, Japan
| | - Akira Naganuma
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Gi-Wook Hwang
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan.
| |
Collapse
|
23
|
Schweighofer N, Genser B, Maerz W, Kleber ME, Trummer O, Pieber TR, Obermayer-Pietsch B. Intronic Variants in OCT1 are Associated with All-Cause and Cardiovascular Mortality in Metformin Users with Type 2 Diabetes. Diabetes Metab Syndr Obes 2020; 13:2069-2080. [PMID: 32606866 PMCID: PMC7308180 DOI: 10.2147/dmso.s235663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 05/01/2020] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Organic cation transporters (Octs) use cations like endogenous compounds, toxins, and drugs, such as metformin, as substrates. Therefore, these proteins determine the pharmacokinetics and -dynamics of metformin and thus its efficacy. Of note, metformin is today the most commonly used pharmaceutical in the treatment of type 2 diabetes (T2DM) with nevertheless a great variability in clinical response, which attributes to genetic variances. The aim of this study was to determine the influence of intronic OCT1 SNPs on prevalence of all-cause and cardiovascular death. PATIENTS AND METHODS Genotypes of 27 intronic SNPs in OCT1 were investigated in the LURIC study, a prospective cohort of 3316 participants scheduled for coronary angiography. We investigated whether these variants were associated with all-cause and cardiovascular death in 73 individuals with T2DM under metformin therapy, in individuals without diabetes, individuals with T2DM and individuals with T2DM without metformin therapy. RESULTS In a multivariate Cox regression analysis adjusted for classical cardiovascular risk factors, 4 intronic OCT1 SNPs were significantly associated with all-cause and cardiovascular mortality in individuals with T2DM on metformin therapy. CONCLUSION According to their OCT1 genotype, some individuals with T2DM on metformin therapy might be prone to an increased risk of cardiovascular death.
Collapse
Affiliation(s)
- Natascha Schweighofer
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University Graz, Graz, Austria
- CBmed GmbH, Center for Biomarker Research in Medicine, Graz, Austria
| | - Bernd Genser
- BG Stats Consulting, Vienna, Austria
- Institute of Public Health, Social and Preventive Medicine, Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
| | - Winfried Maerz
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SynLaboratory Academy, SynLaboratory Holding Deutschland GmbH, Mannheim and Augsburg, Germany
| | - Marcus E Kleber
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Olivia Trummer
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University Graz, Graz, Austria
| | - Thomas R Pieber
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University Graz, Graz, Austria
- CBmed GmbH, Center for Biomarker Research in Medicine, Graz, Austria
| | - Barbara Obermayer-Pietsch
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University Graz, Graz, Austria
- CBmed GmbH, Center for Biomarker Research in Medicine, Graz, Austria
| |
Collapse
|
24
|
Herzog C, Greenald D, Larraz J, Keatinge M, Herrgen L. RNA-seq analysis and compound screening highlight multiple signalling pathways regulating secondary cell death after acute CNS injury in vivo. Biol Open 2020; 9:9/5/bio050260. [PMID: 32366533 PMCID: PMC7225090 DOI: 10.1242/bio.050260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Understanding the molecular mechanisms that regulate secondary cell death after acute central nervous system (CNS) injury is critical for the development of effective neuroprotective drugs. Previous research has shown that neurotoxic processes including excitotoxicity, oxidative stress and neuroinflammation can cause secondary cell death. Nevertheless, clinical trials targeting these processes have been largely unsuccessful, suggesting that the signalling pathways underlying secondary cell death remain incompletely understood. Due to their suitability for live imaging and their amenability to genetic and pharmacological manipulation, larval zebrafish provide an ideal platform for studying the regulation of secondary cell death in vivo Here, we use RNA-seq gene expression profiling and compound screening to identify signalling pathways that regulate secondary cell death after acute neural injury in larval zebrafish. RNA-seq analysis of genes upregulated in cephalic mpeg1+ macrophage-lineage cells isolated from mpeg1:GFP transgenic larvae after neural injury suggested an involvement of cytokine and polyamine signalling in secondary cell death. Furthermore, screening a library of FDA approved compounds indicated roles for GABA, serotonin and dopamine signalling. Overall, our results highlight multiple signalling pathways that regulate secondary cell death in vivo, and thus provide a starting point for the development of novel neuroprotective treatments for patients with CNS injury.This article has an associated First Person interview with the two first authors of the paper.
Collapse
Affiliation(s)
- Chiara Herzog
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - David Greenald
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Juan Larraz
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Marcus Keatinge
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Leah Herrgen
- Centre for Discovery Brain Sciences, Deanery of Biomedical Sciences, The University of Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
25
|
Levasseur EM, Yamada K, Piñeros AR, Wu W, Syed F, Orr KS, Anderson-Baucum E, Mastracci TL, Maier B, Mosley AL, Liu Y, Bernal-Mizrachi E, Alonso LC, Scott D, Garcia-Ocaña A, Tersey SA, Mirmira RG. Hypusine biosynthesis in β cells links polyamine metabolism to facultative cellular proliferation to maintain glucose homeostasis. Sci Signal 2019; 12:eaax0715. [PMID: 31796630 PMCID: PMC7202401 DOI: 10.1126/scisignal.aax0715] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Deoxyhypusine synthase (DHPS) uses the polyamine spermidine to catalyze the hypusine modification of the mRNA translation factor eIF5A and promotes oncogenesis through poorly defined mechanisms. Because germline deletion of Dhps is embryonically lethal, its role in normal postnatal cellular function in vivo remains unknown. We generated a mouse model that enabled the inducible, postnatal deletion of Dhps specifically in postnatal islet β cells, which function to maintain glucose homeostasis. Removal of Dhps did not have an effect under normal physiologic conditions. However, upon development of insulin resistance, which induces β cell proliferation, Dhps deletion caused alterations in proteins required for mRNA translation and protein secretion, reduced production of the cell cycle molecule cyclin D2, impaired β cell proliferation, and induced overt diabetes. We found that hypusine biosynthesis was downstream of protein kinase C-ζ and was required for c-Myc-induced proliferation. Our studies reveal a requirement for DHPS in β cells to link polyamines to mRNA translation to effect facultative cellular proliferation and glucose homeostasis.
Collapse
Affiliation(s)
- Esther M Levasseur
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kentaro Yamada
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Annie R Piñeros
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Wenting Wu
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Farooq Syed
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kara S Orr
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Teresa L Mastracci
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| | - Bernhard Maier
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Amber L Mosley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Laura C Alonso
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Donald Scott
- Diabetes, Obesity, and Metabolism Institute and the Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adolfo Garcia-Ocaña
- Diabetes, Obesity, and Metabolism Institute and the Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sarah A Tersey
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Raghavendra G Mirmira
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
26
|
Ganapathi M, Padgett LR, Yamada K, Devinsky O, Willaert R, Person R, Au PYB, Tagoe J, McDonald M, Karlowicz D, Wolf B, Lee J, Shen Y, Okur V, Deng L, LeDuc CA, Wang J, Hanner A, Mirmira RG, Park MH, Mastracci TL, Chung WK. Recessive Rare Variants in Deoxyhypusine Synthase, an Enzyme Involved in the Synthesis of Hypusine, Are Associated with a Neurodevelopmental Disorder. Am J Hum Genet 2019; 104:287-298. [PMID: 30661771 PMCID: PMC6369575 DOI: 10.1016/j.ajhg.2018.12.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 12/20/2018] [Indexed: 01/23/2023] Open
Abstract
Hypusine is formed post-translationally from lysine and is found in a single cellular protein, eukaryotic translation initiation factor-5A (eIF5A), and its homolog eIF5A2. Biosynthesis of hypusine is a two-step reaction involving the enzymes deoxyhypusine synthase (DHPS) and deoxyhypusine hydroxylase (DOHH). eIF5A is highly conserved throughout eukaryotic evolution and plays a role in mRNA translation, cellular proliferation, cellular differentiation, and inflammation. DHPS is also highly conserved and is essential for life, as Dhps-null mice are embryonic lethal. Using exome sequencing, we identified rare biallelic, recurrent, predicted likely pathogenic variants in DHPS segregating with disease in five affected individuals from four unrelated families. These individuals have similar neurodevelopmental features that include global developmental delay and seizures. Two of four affected females have short stature. All five affected individuals share a recurrent missense variant (c.518A>G [p.Asn173Ser]) in trans with a likely gene disrupting variant (c.1014+1G>A, c.912_917delTTACAT [p.Tyr305_Ile306del], or c.1A>G [p.Met1?]). cDNA studies demonstrated that the c.1014+1G>A variant causes aberrant splicing. Recombinant DHPS enzyme harboring either the p.Asn173Ser or p.Tyr305_Ile306del variant showed reduced (20%) or absent in vitro activity, respectively. We co-transfected constructs overexpressing HA-tagged DHPS (wild-type or mutant) and GFP-tagged eIF5A into HEK293T cells to determine the effect of these variants on hypusine biosynthesis and observed that the p.Tyr305_Ile306del and p.Asn173Ser variants resulted in reduced hypusination of eIF5A compared to wild-type DHPS enzyme. Our data suggest that rare biallelic variants in DHPS result in reduced enzyme activity that limits the hypusination of eIF5A and are associated with a neurodevelopmental disorder.
Collapse
Affiliation(s)
- Mythily Ganapathi
- Personalized Genomic Medicine, Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Leah R Padgett
- Regenerative Medicine & Metabolic Biology, Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| | - Kentaro Yamada
- Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Orrin Devinsky
- Neurology Department, NYU Langone Medical Center, New York, NY 10016, USA
| | | | | | - Ping-Yee Billie Au
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Julia Tagoe
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Marie McDonald
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Danielle Karlowicz
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Barry Wolf
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; Department of Pediatrics, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Joanna Lee
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; Department of Pediatrics, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Volkan Okur
- Department of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
| | - Liyong Deng
- Department of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
| | - Charles A LeDuc
- Department of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
| | - Jiayao Wang
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Ashleigh Hanner
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4340, USA
| | - Raghavendra G Mirmira
- Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Myung Hee Park
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4340, USA
| | - Teresa L Mastracci
- Regenerative Medicine & Metabolic Biology, Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
27
|
Kang B, Jiang D, Ma R, He H, Yi Z, Chen Z. OAZ1 knockdown enhances viability and inhibits ER and LHR transcriptions of granulosa cells in geese. PLoS One 2017; 12:e0175016. [PMID: 28362829 PMCID: PMC5376318 DOI: 10.1371/journal.pone.0175016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/20/2017] [Indexed: 11/18/2022] Open
Abstract
An increasing number of studies suggest that ornithine decarboxylase antizyme 1 (OAZ1), which is regarded as a tumor suppressor gene, regulates follicular development, ovulation, and steroidogenesis. The granulosa cells in the ovary play a critical role in these ovarian functions. However, the action of OAZ1 mediating physiological functions of granulosa cells is obscure. OAZ1 knockdown in granulosa cells of geese was carried out in the current study. The effect of OAZ1 knockdown on polyamine metabolism, cell proliferation, apoptosis, and hormone receptor transcription of primary granulosa cells in geese was measured. The viability of granulosa cells transfected with the shRNA OAZ1 at 48 h was significantly higher than the control (p<0.05). The level of putrescine and spermidine in granulosa cells down-regulating OAZ1 was 7.04- and 2.11- fold higher compared with the control, respectively (p<0.05). The CCND1, SMAD1, and BCL-2 mRNA expression levels in granulosa cells down-regulating OAZ1 were each significantly higher than the control, respectively (p<0.05), whereas the PCNA and CASPASE 3 expression levels were significantly lower than the control (p<0.05). The estradiol concentration, ER and LHR mRNA expression levels were significantly lower in granulosa cells down-regulating OAZ1 compared with the control (p<0.05). Taken together, our results indicated that OAZ1 knockdown elevated the putrescine and spermidine contents and enhanced granulosa cell viability and inhibited ER and LHR transcriptions of granulosa cells in geese.
Collapse
Affiliation(s)
- Bo Kang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan Province, People’s Republic of China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, People’s Republic of China
- * E-mail: (BK); (DMJ)
| | - Dongmei Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan Province, People’s Republic of China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan Province, People’s Republic of China
- * E-mail: (BK); (DMJ)
| | - Rong Ma
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan Province, People’s Republic of China
| | - Hui He
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan Province, People’s Republic of China
| | - Zhixin Yi
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan Province, People’s Republic of China
| | - Ziyu Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan Province, People’s Republic of China
| |
Collapse
|
28
|
Frohnhöfer HG, Geiger-Rudolph S, Pattky M, Meixner M, Huhn C, Maischein HM, Geisler R, Gehring I, Maderspacher F, Nüsslein-Volhard C, Irion U. Spermidine, but not spermine, is essential for pigment pattern formation in zebrafish. Biol Open 2016; 5:736-44. [PMID: 27215328 PMCID: PMC4920196 DOI: 10.1242/bio.018721] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Polyamines are small poly-cations essential for all cellular life. The main polyamines present in metazoans are putrescine, spermidine and spermine. Their exact functions are still largely unclear; however, they are involved in a wide variety of processes affecting cell growth, proliferation, apoptosis and aging. Here we identify idefix, a mutation in the zebrafish gene encoding the enzyme spermidine synthase, leading to a severe reduction in spermidine levels as shown by capillary electrophoresis-mass spectrometry. We show that spermidine, but not spermine, is essential for early development, organogenesis and colour pattern formation. Whereas in other vertebrates spermidine deficiency leads to very early embryonic lethality, maternally provided spermidine synthase in zebrafish is sufficient to rescue the early developmental defects. This allows us to uncouple them from events occurring later during colour patterning. Factors involved in the cellular interactions essential for colour patterning, likely targets for spermidine, are the gap junction components Cx41.8, Cx39.4, and Kir7.1, an inwardly rectifying potassium channel, all known to be regulated by polyamines. Thus, zebrafish provide a vertebrate model to study the in vivo effects of polyamines. Summary: We show that the polyamine spermidine, but not spermine, in addition to more general functions during early development, also specifically regulates colour pattern formation in adult zebrafish.
Collapse
Affiliation(s)
- Hans Georg Frohnhöfer
- Max-Planck-Institut für Entwicklungsbiologie, Abteilung 3, Spemannstrasse 35, Tübingen 72076, Germany
| | - Silke Geiger-Rudolph
- Max-Planck-Institut für Entwicklungsbiologie, Abteilung 3, Spemannstrasse 35, Tübingen 72076, Germany
| | - Martin Pattky
- Institut für Physikalische und Theoretische Chemie, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 18, Tübingen 72076, Germany
| | - Martin Meixner
- Institut für Physikalische und Theoretische Chemie, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 18, Tübingen 72076, Germany
| | - Carolin Huhn
- Institut für Physikalische und Theoretische Chemie, Eberhard Karls Universität Tübingen, Auf der Morgenstelle 18, Tübingen 72076, Germany
| | - Hans-Martin Maischein
- Max-Planck-Institut für Entwicklungsbiologie, Abteilung 3, Spemannstrasse 35, Tübingen 72076, Germany
| | - Robert Geisler
- Max-Planck-Institut für Entwicklungsbiologie, Abteilung 3, Spemannstrasse 35, Tübingen 72076, Germany
| | - Ines Gehring
- Max-Planck-Institut für Entwicklungsbiologie, Abteilung 3, Spemannstrasse 35, Tübingen 72076, Germany
| | - Florian Maderspacher
- Max-Planck-Institut für Entwicklungsbiologie, Abteilung 3, Spemannstrasse 35, Tübingen 72076, Germany
| | | | - Uwe Irion
- Max-Planck-Institut für Entwicklungsbiologie, Abteilung 3, Spemannstrasse 35, Tübingen 72076, Germany
| |
Collapse
|
29
|
Silva TM, Fiuza SM, Marques MPM, Batista de Carvalho LAE, Amado AM. Conformational study and reassessment of the vibrational assignments for Norspermidine. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2016; 157:227-237. [PMID: 26774814 DOI: 10.1016/j.saa.2016.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 01/04/2016] [Indexed: 06/05/2023]
Abstract
The present study presents and discusses the conformational preferences of Norspermidine (NSpd). The effects of varying the dielectric constant on the conformational preferences are discussed, with a view to infer which conformation will correspond to the most stable in the pure condensed liquid phase. Within the same context, a set of NSpd-NH3 molecular adducts were simulated in order to determine the relevance of intermolecular hydrogen bonding on the overall stability and relative positioning of the respective vibrational frequencies. The calculations presently performed allowed a reassessment of the vibrational assignments for NSpd. A full assignment of the NSpd vibrational spectra is presented, with special emphasis being given to the vibrational modes that proved to be most affected by hydrogen bonding. The various inconsistencies of a prior study found in the literature were identified and rectified.
Collapse
Affiliation(s)
- T M Silva
- I&D Unit "Química-Física Molecular", Department of Chemistry, Faculty of Science and Technology, University of Coimbra, P-3004-535 Coimbra, Portugal
| | - S M Fiuza
- I&D Unit "Química-Física Molecular", Department of Chemistry, Faculty of Science and Technology, University of Coimbra, P-3004-535 Coimbra, Portugal
| | - M P M Marques
- I&D Unit "Química-Física Molecular", Department of Chemistry, Faculty of Science and Technology, University of Coimbra, P-3004-535 Coimbra, Portugal; Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
| | - L A E Batista de Carvalho
- I&D Unit "Química-Física Molecular", Department of Chemistry, Faculty of Science and Technology, University of Coimbra, P-3004-535 Coimbra, Portugal
| | - A M Amado
- I&D Unit "Química-Física Molecular", Department of Chemistry, Faculty of Science and Technology, University of Coimbra, P-3004-535 Coimbra, Portugal.
| |
Collapse
|
30
|
Cheng WH, Wang FL, Cheng XQ, Zhu QH, Sun YQ, Zhu HG, Sun J. Polyamine and Its Metabolite H2O2 Play a Key Role in the Conversion of Embryogenic Callus into Somatic Embryos in Upland Cotton (Gossypium hirsutum L.). FRONTIERS IN PLANT SCIENCE 2015; 6:1063. [PMID: 26697030 PMCID: PMC4667013 DOI: 10.3389/fpls.2015.01063] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/16/2015] [Indexed: 05/23/2023]
Abstract
The objective of this study was to increase understanding about the mechanism by which polyamines (PAs) promote the conversion of embryogenic calli (EC) into somatic embryos in cotton (Gossypium hirsutum L.). We measured the levels of endogenous PAs and H2O2, quantified the expression levels of genes involved in the PAs pathway at various stages of cotton somatic embryogenesis (SE), and investigated the effects of exogenous PAs and H2O2 on differentiation and development of EC. Putrescine (Put), spermidine (Spd), and spermine (Spm) significantly increased from the EC stage to the early phase of embryo differentiation. The levels of Put then decreased until the somatic embryo stage whereas Spd and Spm remained nearly the same. The expression profiles of GhADC genes were consistent with changes in Put during cotton SE. The H2O2 concentrations began to increase significantly at the EC stage, during which time both GhPAO1 and GhPAO4 expressions were highest and PAO activity was significantly increased. Exogenous Put, Spd, Spm, and H2O2 not only enhanced embryogenic callus growth and embryo formation, but also alleviated the effects of D-arginine and 1, 8-diamino-octane, which are inhibitors of PA synthesis and PAO activity. Overall, the results suggest that both PAs and their metabolic product H2O2 are essential for the conversion of EC into somatic embryos in cotton.
Collapse
Affiliation(s)
- Wen-Han Cheng
- College of Agriculture/The Key Laboratory of Oasis Eco-Agriculture, Shihezi UniversityShihezi, China
| | - Fan-Long Wang
- College of Agriculture/The Key Laboratory of Oasis Eco-Agriculture, Shihezi UniversityShihezi, China
| | - Xin-Qi Cheng
- College of Agriculture/The Key Laboratory of Oasis Eco-Agriculture, Shihezi UniversityShihezi, China
| | - Qian-Hao Zhu
- Agriculture, Commonwealth Scientific and Industrial Research OrganisationCanberra, ACT, Australia
| | - Yu-Qiang Sun
- College of Agriculture/The Key Laboratory of Oasis Eco-Agriculture, Shihezi UniversityShihezi, China
- College of Life and Environmental Science, Hangzhou Normal UniversityHangzhou, China
| | - Hua-Guo Zhu
- College of Agriculture/The Key Laboratory of Oasis Eco-Agriculture, Shihezi UniversityShihezi, China
- *Correspondence: Jie Sun, ; Hua-Guo Zhu,
| | - Jie Sun
- College of Agriculture/The Key Laboratory of Oasis Eco-Agriculture, Shihezi UniversityShihezi, China
- *Correspondence: Jie Sun, ; Hua-Guo Zhu,
| |
Collapse
|