1
|
Salagre D, Bajit H, Fernández-Vázquez G, Dwairy M, Garzón I, Haro-López R, Agil A. Melatonin induces fiber switching by improvement of mitochondrial oxidative capacity and function via NRF2/RCAN/MEF2 in the vastus lateralis muscle from both sex Zücker diabetic fatty rats. Free Radic Biol Med 2024; 227:322-335. [PMID: 39645208 DOI: 10.1016/j.freeradbiomed.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
The positive role of melatonin in obesity control and skeletal muscle (SKM) preservation is well known. We recently showed that melatonin improves vastus lateralis muscle (VL) fiber oxidative phenotype. However, fiber type characterization, mitochondrial function, and molecular mechanisms that underlie VL fiber switching by melatonin are still undefined. Our study aims to investigate whether melatonin induces fiber switching by NRF2/RCAN/MEF2 pathway activation and mitochondrial oxidative metabolism modulation in the VL of both sex Zücker diabetic fatty (ZDF) rats. 5-Weeks-old male and female ZDF rats (N = 16) and their age-matched lean littermates (ZL) were subdivided into two subgroups: control (C) and orally treated with melatonin (M) (10 mg/kg/day) for 12 weeks. Interestingly, melatonin increased oxidative fibers amounts (Types I and IIa) counteracting the decreased levels found in the VL of obese-diabetic rats, and upregulated NRF2, calcineurin and MEF2 expression. Melatonin also restored the mitochondrial oxidative capacity increasing the respiratory control ratio (RCR) in both sex and phenotype rats through the reduction of the proton leak component of respiration (state 4). Melatonin also improved the VL mitochondrial phosphorylation coefficient and modulated the total oxygen consumption by enhancing complex I, III and IV activity, and fatty acid oxidation (FAO) in both sex obese-diabetic rats, decreasing in male and increasing in female the complex II oxygen consumption. These findings suggest that melatonin treatment induces fiber switching in SKM improving mitochondrial functionality by NRF2/RCAN/MEF2 pathway activation.
Collapse
Affiliation(s)
- Diego Salagre
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, 18016, Granada, Spain
| | - Habiba Bajit
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, 18016, Granada, Spain
| | | | - Mutaz Dwairy
- Department of Civil Engineering, Yarmuk University, 21163, Irbid, Jordan
| | - Ingrid Garzón
- Tissue Engineering Group, Department of Histology, BioHealth Institute Granada (IBs Granada), School of Medicine, University of Granada, 18016, Granada, Spain
| | - Rocío Haro-López
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, 18016, Granada, Spain
| | - Ahmad Agil
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, 18016, Granada, Spain.
| |
Collapse
|
2
|
Tallino S, Etebari R, McDonough I, Leon H, Sepulveda I, Winslow W, Bartholomew SK, Perez SE, Mufson EJ, Velazquez R. Assessing the Benefit of Dietary Choline Supplementation Throughout Adulthood in the Ts65Dn Mouse Model of Down Syndrome. Nutrients 2024; 16:4167. [PMID: 39683562 DOI: 10.3390/nu16234167] [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: 11/06/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Down syndrome (DS) is the most common cause of early-onset Alzheimer's disease (AD). Dietary choline has been proposed as a modifiable factor to improve the cognitive and pathological outcomes of AD and DS, especially as many do not reach adequate daily intake levels of choline. While lower circulating choline levels correlate with worse pathological measures in AD patients, choline status and intake in DS is widely understudied. Perinatal choline supplementation (Ch+) in the Ts65Dn mouse model of DS protects offspring against AD-relevant pathology and improves cognition. Further, dietary Ch+ in adult AD models also ameliorates pathology and improves cognition. However, dietary Ch+ in adult Ts65Dn mice has not yet been explored; thus, this study aimed to supply Ch+ throughout adulthood to determine the effects on cognition and DS co-morbidities. METHODS We fed trisomic Ts65Dn mice and disomic littermate controls either a choline normal (ChN; 1.1 g/kg) or a Ch+ (5 g/kg) diet from 4.5 to 14 months of age. RESULTS We found that Ch+ in adulthood failed to improve genotype-specific deficits in spatial learning. However, in both genotypes of female mice, Ch+ significantly improved cognitive flexibility in a reverse place preference task in the IntelliCage behavioral phenotyping system. Further, Ch+ significantly reduced weight gain and peripheral inflammation in female mice of both genotypes, and significantly improved glucose metabolism in male mice of both genotypes. CONCLUSIONS Our findings suggest that adulthood choline supplementation benefits behavioral and biological factors important for general well-being in DS and related to AD risk.
Collapse
Affiliation(s)
- Savannah Tallino
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Rachel Etebari
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Ian McDonough
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Hector Leon
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Isabella Sepulveda
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Wendy Winslow
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Samantha K Bartholomew
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Sylvia E Perez
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Elliott J Mufson
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Ramon Velazquez
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| |
Collapse
|
3
|
Hushmandi K, Einollahi B, Aow R, Suhairi SB, Klionsky DJ, Aref AR, Reiter RJ, Makvandi P, Rabiee N, Xu Y, Nabavi N, Saadat SH, Farahani N, Kumar AP. Investigating the interplay between mitophagy and diabetic neuropathy: Uncovering the hidden secrets of the disease pathology. Pharmacol Res 2024; 208:107394. [PMID: 39233055 DOI: 10.1016/j.phrs.2024.107394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/18/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Mitophagy, the cellular process of selectively eliminating damaged mitochondria, plays a crucial role in maintaining metabolic balance and preventing insulin resistance, both key factors in type 2 diabetes mellitus (T2DM) development. When mitophagy malfunctions in diabetic neuropathy, it triggers a cascade of metabolic disruptions, including reduced energy production, increased oxidative stress, and cell death, ultimately leading to various complications. Thus, targeting mitophagy to enhance the process may have emerged as a promising therapeutic strategy for T2DM and its complications. Notably, plant-derived compounds with β-cell protective and mitophagy-stimulating properties offer potential as novel therapeutic agents. This review highlights the intricate mechanisms linking mitophagy dysfunction to T2DM and its complications, particularly neuropathy, elucidating potential therapeutic interventions for this debilitating disease.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Rachel Aow
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suhana Binte Suhairi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Amir Reza Aref
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | - Pooyan Makvandi
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India; University Centre for Research & Development, Chandigarh University, Mohali, Punjab 140413, India
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India
| | - Yi Xu
- Department of Science & Technology, Department of Urology, NanoBioMed Group, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
4
|
Zhakula-Kostadinova N, Taylor AM. Patterns of Aneuploidy and Signaling Consequences in Cancer. Cancer Res 2024; 84:2575-2587. [PMID: 38924459 PMCID: PMC11325152 DOI: 10.1158/0008-5472.can-24-0169] [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: 01/16/2024] [Revised: 03/29/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Aneuploidy, or a change in the number of whole chromosomes or chromosome arms, is a near-universal feature of cancer. Chromosomes affected by aneuploidy are not random, with observed cancer-specific and tissue-specific patterns. Recent advances in genome engineering methods have allowed the creation of models with targeted aneuploidy events. These models can be used to uncover the downstream effects of individual aneuploidies on cancer phenotypes including proliferation, apoptosis, metabolism, and immune signaling. Here, we review the current state of research into the patterns of aneuploidy in cancer and their impact on signaling pathways and biological processes.
Collapse
Affiliation(s)
- Nadja Zhakula-Kostadinova
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| | - Alison M Taylor
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| |
Collapse
|
5
|
Gruszka J, Włodarek D. General Dietary Recommendations for People with Down Syndrome. Nutrients 2024; 16:2656. [PMID: 39203792 PMCID: PMC11357503 DOI: 10.3390/nu16162656] [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: 07/02/2024] [Revised: 07/24/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Down syndrome (DS) is caused by trisomy of chromosome 21 and is associated with characteristic features of appearance, intellectual impairment to varying degrees, organ defects, and health problems typical of this syndrome. Studies on the frequency of consumption of food products in this group show many irregularities, in particular too low consumption of vegetables and fruits, wholegrain cereal products and dairy products, and excessive consumption of meat products and sweets. It is necessary to correct eating habits. The diets of people with trisomy 21 should be consistent with the recommendations of rational nutrition for the general population and take into account specific dietary modifications related to the occurrence of diseases and health problems characteristic of this syndrome.
Collapse
Affiliation(s)
- Joanna Gruszka
- Dieta Plus Nutritional and Dietary Counseling, 45-072 Opole, Poland;
| | - Dariusz Włodarek
- Department of Dietetics, Institute of Human Nutrition Science, Warsaw University of Life Science (WULS-SGGW), 159C Nowoursynowska Street, 02-776 Warszawa, Poland
| |
Collapse
|
6
|
Lee J, Cho K, Weigel KA, White HM, Do C, Choi I. Identification of genomic regions and genes associated with subclinical ketosis in periparturient dairy cows. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2024; 66:567-576. [PMID: 38975580 PMCID: PMC11222121 DOI: 10.5187/jast.2023.e97] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 07/09/2024]
Abstract
Subclinical ketosis (SCK) is a prevalent metabolic disorder that occurs during the transition to lactation period. It is defined as a high blood concentration of ketone bodies (beta-hydroxybutyric acid f ≥ 1.2 mmol/L) within the first few weeks of lactation, and often presents without clinical signs. SCK is mainly caused by negative energy balance (NEB). The objective of this study is to identify single nucleotide polymorphisms (SNPs) associated with SCK using genome-wide association studies (GWAS), and to predict the biological functions of proximal genes using gene-set enrichment analysis (GSEA). Blood samples were collected from 112 Holstein cows between 5 and 18 days postpartum to determine the incidence of SCK. Genomic DNA extracted from both SCK and healthy cows was examined using the Illumina Bovine SNP50K BeadChip for genotyping. GWAS revealed 194 putative SNPs and 163 genes associated with those SNPs. Additionally, GSEA showed that the genes retrieved by Database for Annotation, Visualization, and Integrated Discovery (DAVID) belonged to calcium signaling, starch and sucrose, immune network, and metabolic pathways. Furthermore, the proximal genes were found to be related to germ cell and early embryo development. In summary, this study proposes several feasible SNPs and genes associated with SCK through GWAS and GSEA. These candidates can be utilized in selective breeding programs to reduce the genetic risk for SCK and subfertility in high-performance dairy cows.
Collapse
Affiliation(s)
- Jihwan Lee
- Dairy Science Division, National Institute
of Animal Science, RDA, Cheon 31000, Korea
| | - KwangHyeon Cho
- Department of Beef and Dairy Science,
Korea National College of Agriculture and Fisheries, Jeonju
54874, Korea
| | - Kent A. Weigel
- Department of Animal and Dairy Sciences,
University of Wisconsin, Madison 53706, USA
| | - Heather M. White
- Department of Animal and Dairy Sciences,
University of Wisconsin, Madison 53706, USA
| | - ChangHee Do
- Department of Animal and Dairy Sciences,
Chungnam National University, Daejeon 34134, Korea
| | - Inchul Choi
- Department of Animal and Dairy Sciences,
Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
7
|
Chapman LR, Ramnarine IVP, Zemke D, Majid A, Bell SM. Gene Expression Studies in Down Syndrome: What Do They Tell Us about Disease Phenotypes? Int J Mol Sci 2024; 25:2968. [PMID: 38474215 DOI: 10.3390/ijms25052968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Down syndrome is a well-studied aneuploidy condition in humans, which is associated with various disease phenotypes including cardiovascular, neurological, haematological and immunological disease processes. This review paper aims to discuss the research conducted on gene expression studies during fetal development. A descriptive review was conducted, encompassing all papers published on the PubMed database between September 1960 and September 2022. We found that in amniotic fluid, certain genes such as COL6A1 and DSCR1 were found to be affected, resulting in phenotypical craniofacial changes. Additionally, other genes such as GSTT1, CLIC6, ITGB2, C21orf67, C21orf86 and RUNX1 were also identified to be affected in the amniotic fluid. In the placenta, dysregulation of genes like MEST, SNF1LK and LOX was observed, which in turn affected nervous system development. In the brain, dysregulation of genes DYRK1A, DNMT3L, DNMT3B, TBX1, olig2 and AQP4 has been shown to contribute to intellectual disability. In the cardiac tissues, dysregulated expression of genes GART, ETS2 and ERG was found to cause abnormalities. Furthermore, dysregulation of XIST, RUNX1, SON, ERG and STAT1 was observed, contributing to myeloproliferative disorders. Understanding the differential expression of genes provides insights into the genetic consequences of DS. A better understanding of these processes could potentially pave the way for the development of genetic and pharmacological therapies.
Collapse
Affiliation(s)
- Laura R Chapman
- Sheffield Children's NHS Foundation Trust, Clarkson St, Sheffield S10 2TH, UK
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Glossop Road, Sheffield S10 2GF, UK
| | - Isabela V P Ramnarine
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Glossop Road, Sheffield S10 2GF, UK
| | - Dan Zemke
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Glossop Road, Sheffield S10 2GF, UK
| | - Arshad Majid
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Glossop Road, Sheffield S10 2GF, UK
- Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2GJ, UK
| | - Simon M Bell
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Glossop Road, Sheffield S10 2GF, UK
- Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2GJ, UK
| |
Collapse
|
8
|
Xing Z, Li Y, Cortes-Gomez E, Jiang X, Gao S, Pao A, Shan J, Song Y, Perez A, Yu T, Highsmith MR, Boadu F, Conroy JM, Singh PK, Bakin AV, Cheng J, Duan Z, Wang J, Liu S, Tycko B, Yu YE. Dissection of a Down syndrome-associated trisomy to separate the gene dosage-dependent and -independent effects of an extra chromosome. Hum Mol Genet 2023; 32:2205-2218. [PMID: 37014740 PMCID: PMC10281752 DOI: 10.1093/hmg/ddad056] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/13/2023] [Accepted: 03/27/2023] [Indexed: 04/05/2023] Open
Abstract
As an aneuploidy, trisomy is associated with mammalian embryonic and postnatal abnormalities. Understanding the underlying mechanisms involved in mutant phenotypes is broadly important and may lead to new strategies to treat clinical manifestations in individuals with trisomies, such as trisomy 21 [Down syndrome (DS)]. Although increased gene dosage effects because of a trisomy may account for the mutant phenotypes, there is also the possibility that phenotypic consequences of a trisomy can arise because of the presence of a freely segregating extra chromosome with its own centromere, i.e. a 'free trisomy' independent of gene dosage effects. Presently, there are no reports of attempts to functionally separate these two types of effects in mammals. To fill this gap, here we describe a strategy that employed two new mouse models of DS, Ts65Dn;Df(17)2Yey/+ and Dp(16)1Yey/Df(16)8Yey. Both models carry triplications of the same 103 human chromosome 21 gene orthologs; however, only Ts65Dn;Df(17)2Yey/+ mice carry a free trisomy. Comparison of these models revealed the gene dosage-independent impacts of an extra chromosome at the phenotypic and molecular levels for the first time. They are reflected by impairments of Ts65Dn;Df(17)2Yey/+ males in T-maze tests when compared with Dp(16)1Yey/Df(16)8Yey males. Results from the transcriptomic analysis suggest the extra chromosome plays a major role in trisomy-associated expression alterations of disomic genes beyond gene dosage effects. This model system can now be used to deepen our mechanistic understanding of this common human aneuploidy and obtain new insights into the effects of free trisomies in other human diseases such as cancers.
Collapse
Affiliation(s)
- Zhuo Xing
- The Children’s Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Yichen Li
- The Children’s Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Eduardo Cortes-Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Xiaoling Jiang
- The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Shuang Gao
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Bioinformatics, OmniSeq Inc., Buffalo, NY, USA
| | - Annie Pao
- The Children’s Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jidong Shan
- Molecular Cytogenetics Core, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yinghui Song
- Molecular Cytogenetics Core, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Amanda Perez
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Tao Yu
- The Children’s Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Max R Highsmith
- Department of Electric Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Frimpong Boadu
- Department of Electric Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Jeffrey M Conroy
- Research and Development, OmniSeq Inc., Buffalo, NY, USA
- Research Support Services, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Prashant K Singh
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Andrei V Bakin
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jianlin Cheng
- Department of Electric Engineering and Computer Science, University of Missouri, Columbia, MO, USA
| | - Zhijun Duan
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Benjamin Tycko
- Hackensack-Meridian Health Center for Discovery and Innovation, Nutley, NJ, USA
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Y Eugene Yu
- The Children’s Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Genetics, Genomics and Bioinformatics Program, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
9
|
Sarver DC, Xu C, Velez LM, Aja S, Jaffe AE, Seldin MM, Reeves RH, Wong GW. Dysregulated systemic metabolism in a Down syndrome mouse model. Mol Metab 2023; 68:101666. [PMID: 36587842 PMCID: PMC9841171 DOI: 10.1016/j.molmet.2022.101666] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/14/2022] [Accepted: 12/26/2022] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Trisomy 21 is one of the most complex genetic perturbations compatible with postnatal survival. Dosage imbalance arising from the triplication of genes on human chromosome 21 (Hsa21) affects multiple organ systems. Much of Down syndrome (DS) research, however, has focused on addressing how aneuploidy dysregulates CNS function leading to cognitive deficit. Although obesity, diabetes, and associated sequelae such as fatty liver and dyslipidemia are well documented in the DS population, only limited studies have been conducted to determine how gene dosage imbalance affects whole-body metabolism. Here, we conduct a comprehensive and systematic analysis of key metabolic parameters across different physiological states in the Ts65Dn trisomic mouse model of DS. METHODS Ts65Dn mice and euploid littermates were subjected to comprehensive metabolic phenotyping under basal (chow-fed) state and the pathophysiological state of obesity induced by a high-fat diet (HFD). RNA sequencing of liver, skeletal muscle, and two major fat depots were conducted to determine the impact of aneuploidy on tissue transcriptome. Pathway enrichments, gene-centrality, and key driver estimates were performed to provide insights into tissue autonomous and non-autonomous mechanisms contributing to the dysregulation of systemic metabolism. RESULTS Under the basal state, chow-fed Ts65Dn mice of both sexes had elevated locomotor activity and energy expenditure, reduced fasting serum cholesterol levels, and mild glucose intolerance. Sexually dimorphic deterioration in metabolic homeostasis became apparent when mice were challenged with a high-fat diet. While obese Ts65Dn mice of both sexes exhibited dyslipidemia, male mice also showed impaired systemic insulin sensitivity, reduced mitochondrial activity, and elevated fibrotic and inflammatory gene signatures in the liver and adipose tissue. Systems-level analysis highlighted conserved pathways and potential endocrine drivers of adipose-liver crosstalk that contribute to dysregulated glucose and lipid metabolism. CONCLUSIONS A combined alteration in the expression of trisomic and disomic genes in peripheral tissues contribute to metabolic dysregulations in Ts65Dn mice. These data lay the groundwork for understanding the impact of aneuploidy on in vivo metabolism.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cheng Xu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leandro M Velez
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA; Center for Epigenetics and Metabolism, University of California Irvine, Irvine, USA
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew E Jaffe
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; The Lieber Institute for Brain Development, Baltimore, MD, USA; Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA; Center for Epigenetics and Metabolism, University of California Irvine, Irvine, USA
| | - Roger H Reeves
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
10
|
Martini AC, Gross TJ, Head E, Mapstone M. Beyond amyloid: Immune, cerebrovascular, and metabolic contributions to Alzheimer disease in people with Down syndrome. Neuron 2022; 110:2063-2079. [PMID: 35472307 PMCID: PMC9262826 DOI: 10.1016/j.neuron.2022.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 12/16/2022]
Abstract
People with Down syndrome (DS) have increased risk of Alzheimer disease (AD), presumably conferred through genetic predispositions arising from trisomy 21. These predispositions necessarily include triplication of the amyloid precursor protein (APP), but also other Ch21 genes that confer risk directly or through interactions with genes on other chromosomes. We discuss evidence that multiple genes on chromosome 21 are associated with metabolic dysfunction in DS. The resulting dysregulated pathways involve the immune system, leading to chronic inflammation; the cerebrovascular system, leading to disruption of the blood brain barrier (BBB); and cellular energy metabolism, promoting increased oxidative stress. In combination, these disruptions may produce a precarious biological milieu that, in the presence of accumulating amyloid, drives the pathophysiological cascade of AD in people with DS. Critically, mechanistic drivers of this dysfunction may be targetable in future clinical trials of pharmaceutical and/or lifestyle interventions.
Collapse
Affiliation(s)
- Alessandra C Martini
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Thomas J Gross
- Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Mark Mapstone
- Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
11
|
Shan Z, Fa WH, Tian CR, Yuan CS, Jie N. Mitophagy and mitochondrial dynamics in type 2 diabetes mellitus treatment. Aging (Albany NY) 2022; 14:2902-2919. [PMID: 35332108 PMCID: PMC9004550 DOI: 10.18632/aging.203969] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/03/2021] [Indexed: 01/18/2023]
Abstract
The prevalence of type 2 diabetes is associated with inflammatory bowels diseases, nonalcoholic steatohepatitis and even a spectrum of cancer such as colon cancer and liver cancer, resulting in a substantial healthcare burden on our society. Autophagy is a key regulator in metabolic homeostasis such as lipid metabolism, energy management and the balance of cellular mineral substances. Mitophagy is selective autophagy for clearing the damaged mitochondria and dysfunctional mitochondria. A myriad of evidence has demonstrated a major role of mitophagy in the regulation of type 2 diabetes and metabolic homeostasis. It is well established that defective mitophagy has been linked to the development of insulin resistance. Moreover, insulin resistance is further progressed to various diseases such as nephropathy, retinopathy and cardiovascular diseases. Concordantly, restoration of mitophagy will be a reliable and therapeutic target for type 2 diabetes. Recently, various phytochemicals have been proved to prevent dysfunctions of β-cells by mitophagy inductions during diabetes developments. In agreement with the above phenomenon, mitophagy inducers should be warranted as potential and novel therapeutic agents for treating diabetes. This review focuses on the role of mitophagy in type 2 diabetes relevant diseases and the pharmacological basis and therapeutic potential of autophagy regulators in type 2 diabetes.
Collapse
Affiliation(s)
- Zhao Shan
- Department of Endocrinology, Shenzhen Longhua District Central Hospital, Guangdong Medical University Afliated Longhua Central Hospital, Shenzhen 518110, Guangdong, China
| | - Wei Hong Fa
- Department of Endocrinology, Shenzhen Longhua District Central Hospital, Guangdong Medical University Afliated Longhua Central Hospital, Shenzhen 518110, Guangdong, China
| | - Chen Run Tian
- Department of Endocrinology, Shenzhen Longhua District Central Hospital, Guangdong Medical University Afliated Longhua Central Hospital, Shenzhen 518110, Guangdong, China
| | - Chen Shi Yuan
- Department of Endocrinology, Shenzhen Longhua District Central Hospital, Guangdong Medical University Afliated Longhua Central Hospital, Shenzhen 518110, Guangdong, China
| | - Ning Jie
- Department of Endocrinology, Shenzhen Longhua District Central Hospital, Guangdong Medical University Afliated Longhua Central Hospital, Shenzhen 518110, Guangdong, China
| |
Collapse
|
12
|
Venegas-Zamora L, Bravo-Acuña F, Sigcho F, Gomez W, Bustamante-Salazar J, Pedrozo Z, Parra V. New Molecular and Organelle Alterations Linked to Down Syndrome Heart Disease. Front Genet 2022; 12:792231. [PMID: 35126461 PMCID: PMC8808411 DOI: 10.3389/fgene.2021.792231] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/13/2021] [Indexed: 12/13/2022] Open
Abstract
Down syndrome (DS) is a genetic disorder caused by a trisomy of the human chromosome 21 (Hsa21). Overexpression of Hsa21 genes that encode proteins and non-coding RNAs (ncRNAs) can disrupt several cellular functions and biological processes, especially in the heart. Congenital heart defects (CHDs) are present in 45–50% of individuals with DS. Here, we describe the genetic background of this condition (Hsa21 and non-Hsa21 genes), including the role of ncRNAs, and the relevance of these new players in the study of the pathophysiology of DS heart diseases. Additionally, we discuss several distinct pathways in cardiomyocytes which help maintain a functional heart, but that might trigger hypertrophy and oxidative stress when altered. Moreover, we highlight the importance of investigating how mitochondrial and lysosomal dysfunction could eventually contribute to understanding impaired heart function and development in subjects with the Hsa21 trisomy. Altogether, this review focuses on the newest insights about the gene expression, molecular pathways, and organelle alterations involved in the cardiac phenotype of DS.
Collapse
Affiliation(s)
- Leslye Venegas-Zamora
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Francisco Bravo-Acuña
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Francisco Sigcho
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Wileidy Gomez
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Laboratory of Neuroprotection and Autophagy, Center for Integrative Biology, Faculty of Science, Universidad Mayor, Santiago, Chile
| | - José Bustamante-Salazar
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Zully Pedrozo
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Red para El Estudio de Enfermedades Cardiopulmonares de Alta Letalidad (REECPAL), Universidad de Chile, Santiago, Chile
- *Correspondence: Zully Pedrozo, ; Valentina Parra,
| | - Valentina Parra
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- Red para El Estudio de Enfermedades Cardiopulmonares de Alta Letalidad (REECPAL), Universidad de Chile, Santiago, Chile
- *Correspondence: Zully Pedrozo, ; Valentina Parra,
| |
Collapse
|
13
|
Alldred MJ, Lee SH, Ginsberg SD. Adiponectin Modulation by Genotype and Maternal Choline Supplementation in a Mouse Model of Down Syndrome and Alzheimer's Disease. J Clin Med 2021; 10:2994. [PMID: 34279477 PMCID: PMC8267749 DOI: 10.3390/jcm10132994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 12/13/2022] Open
Abstract
Down syndrome (DS) is a genetic disorder caused by the triplication of human chromosome 21, which results in neurological and physiological pathologies. These deficits increase during aging and are exacerbated by cognitive decline and increase of Alzheimer's disease (AD) neuropathology. A nontoxic, noninvasive treatment, maternal choline supplementation (MCS) attenuates cognitive decline in mouse models of DS and AD. To evaluate potential underlying mechanisms, laser capture microdissection of individual neuronal populations of MCS offspring was performed, followed by RNA sequencing and bioinformatic inquiry. Results at ~6 months of age (MO) revealed DS mice (the well-established Ts65Dn model) have significant dysregulation of select genes within the Type 2 Diabetes Mellitus (T2DM) signaling pathway relative to normal disomic (2N) littermates. Accordingly, we interrogated key T2DM protein hormones by ELISA assay in addition to gene and encoded protein levels in the brain. We found dysregulation of adiponectin (APN) protein levels in the frontal cortex of ~6 MO trisomic mice, which was attenuated by MCS. APN receptors also displayed expression level changes in response to MCS. APN is a potential biomarker for AD pathology and may be relevant in DS. We posit that changes in APN signaling may be an early marker of cognitive decline and neurodegeneration.
Collapse
Affiliation(s)
- Melissa J. Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY 10962, USA
- Departments of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sang Han Lee
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, NY 10962, USA;
- Child & Adolescent Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY 10962, USA
- Departments of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
- Neuroscience & Physiology, New York University Grossman School of Medicine, New York, NY 10016, USA
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
14
|
Bayona-Bafaluy MP, Garrido-Pérez N, Meade P, Iglesias E, Jiménez-Salvador I, Montoya J, Martínez-Cué C, Ruiz-Pesini E. Down syndrome is an oxidative phosphorylation disorder. Redox Biol 2021; 41:101871. [PMID: 33540295 PMCID: PMC7859316 DOI: 10.1016/j.redox.2021.101871] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/29/2020] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
Down syndrome is the most common genomic disorder of intellectual disability and is caused by trisomy of chromosome 21. Several genes in this chromosome repress mitochondrial biogenesis. The goal of this study was to evaluate whether early overexpression of these genes may cause a prenatal impairment of oxidative phosphorylation negatively affecting neurogenesis. Reduction in the mitochondrial energy production and a lower mitochondrial function have been reported in diverse tissues or cell types, and also at any age, including early fetuses, suggesting that a defect in oxidative phosphorylation is an early and general event in Down syndrome individuals. Moreover, many of the medical conditions associated with Down syndrome are also frequently found in patients with oxidative phosphorylation disease. Several drugs that enhance mitochondrial biogenesis are nowadays available and some of them have been already tested in mouse models of Down syndrome restoring neurogenesis and cognitive defects. Because neurogenesis relies on a correct mitochondrial function and critical periods of brain development occur mainly in the prenatal and early neonatal stages, therapeutic approaches intended to improve oxidative phosphorylation should be provided in these periods.
Collapse
Affiliation(s)
- M Pilar Bayona-Bafaluy
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Rd de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Universidad de Zaragoza. C/ Mariano Esquillor (Edificio I+D), 50018, Zaragoza, Spain.
| | - Nuria Garrido-Pérez
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Rd de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Universidad de Zaragoza. C/ Mariano Esquillor (Edificio I+D), 50018, Zaragoza, Spain.
| | - Patricia Meade
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Rd de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Universidad de Zaragoza. C/ Mariano Esquillor (Edificio I+D), 50018, Zaragoza, Spain.
| | - Eldris Iglesias
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain.
| | - Irene Jiménez-Salvador
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain.
| | - Julio Montoya
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Rd de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain.
| | - Carmen Martínez-Cué
- Departamento de Fisiología y Farmacología. Facultad de Medicina, Universidad de Cantabria. Av. Herrera Oría, 39011, Santander, Spain.
| | - Eduardo Ruiz-Pesini
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet, 177. 50013, Zaragoza, Spain and C/ Pedro Cerbuna, 12, 50009, Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, Av. San Juan Bosco, 13, 50009, Zaragoza, Spain; Centro de Investigaciones Biomédicas en Rd de Enfermedades Raras (CIBERER), Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain.
| |
Collapse
|
15
|
Hendrix JA, Amon A, Abbeduto L, Agiovlasitis S, Alsaied T, Anderson HA, Bain LJ, Baumer N, Bhattacharyya A, Bogunovic D, Botteron KN, Capone G, Chandan P, Chase I, Chicoine B, Cieuta-Walti C, DeRuisseau LR, Durand S, Esbensen A, Fortea J, Giménez S, Granholm AC, Hahn LJ, Head E, Hillerstrom H, Jacola LM, Janicki MP, Jasien JM, Kamer AR, Kent RD, Khor B, Lawrence JB, Lemonnier C, Lewanda AF, Mobley W, Moore PE, Nelson LP, Oreskovic NM, Osorio RS, Patterson D, Rasmussen SA, Reeves RH, Roizen N, Santoro S, Sherman SL, Talib N, Tapia IE, Walsh KM, Warren SF, White AN, Wong GW, Yi JS. Opportunities, barriers, and recommendations in down syndrome research. TRANSLATIONAL SCIENCE OF RARE DISEASES 2021; 5:99-129. [PMID: 34268067 PMCID: PMC8279178 DOI: 10.3233/trd-200090] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recent advances in medical care have increased life expectancy and improved the quality of life for people with Down syndrome (DS). These advances are the result of both pre-clinical and clinical research but much about DS is still poorly understood. In 2020, the NIH announced their plan to update their DS research plan and requested input from the scientific and advocacy community. OBJECTIVE The National Down Syndrome Society (NDSS) and the LuMind IDSC Foundation worked together with scientific and medical experts to develop recommendations for the NIH research plan. METHODS NDSS and LuMind IDSC assembled over 50 experts across multiple disciplines and organized them in eleven working groups focused on specific issues for people with DS. RESULTS This review article summarizes the research gaps and recommendations that have the potential to improve the health and quality of life for people with DS within the next decade. CONCLUSIONS This review highlights many of the scientific gaps that exist in DS research. Based on these gaps, a multidisciplinary group of DS experts has made recommendations to advance DS research. This paper may also aid policymakers and the DS community to build a comprehensive national DS research strategy.
Collapse
Affiliation(s)
| | - Angelika Amon
- Deceased. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Leonard Abbeduto
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA; MIND Institute, University of California, Davis, CA, USA
| | | | - Tarek Alsaied
- Heart Institute Department of Pediatrics Cincinnati Children’s Hospital Medical Center University of Cincinnati, Cincinnati, OH, USA
| | | | | | - Nicole Baumer
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA; Down Syndrome Program, Developmental Medicine Center, Boston Children’s Hospital, Boston, MA, USA
| | - Anita Bhattacharyya
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Dusan Bogunovic
- Department of Microbiology, Icahn School of Medicine at Mt. Sinai, New York, NY, USA; Department of Pediatrics, Icahn School of Medicine at Mt. Sinai, New York, NY; Precision Immunology Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Kelly N. Botteron
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Priya Chandan
- Department of Neurosurgery, Division of Physical Medicine and Rehabilitation, University of Louisville School of Medicine, Louisville, KY, USA
| | - Isabelle Chase
- Department of Pediatric Dentistry, Boston Children’s Hospital, Boston, MA, USA
| | - Brian Chicoine
- Advocate Medical Group Adult Down Syndrome Center, Park Ridge, IL, USA
| | | | | | | | - Anna Esbensen
- Department of Pediatrics, University of Cincinnati College of Medicine & Division of Developmental and Behavioral Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Juan Fortea
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain; Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Center of Biomedical Investigation Network for Neurodegenerative Diseases, Madrid, Spain
| | - Sandra Giménez
- Multidisciplinary Sleep Unit, Respiratory Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ann-Charlotte Granholm
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - Laura J. Hahn
- Department of Speech and Hearing Science, University of Illinois Urbana Champaign, Champaign, IL, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, UC Irvine School of Medicine, Orange, CA, USA
| | | | - Lisa M. Jacola
- Department of Psychology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | | | - Joan M. Jasien
- Division of Pediatric Neurology, Duke University Health System, Durham, NC, USA
| | - Angela R. Kamer
- Department of Periodontology and Implant Dentistry, New York University, College of Dentistry, New York, NY, USA
| | - Raymond D. Kent
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Bernard Khor
- Benaroy Research Institute at Virginia Mason, Seattle, WA, USA
| | - Jeanne B. Lawrence
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA; Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Amy Feldman Lewanda
- Children s National Rare Disease Institute, Children’s National Health System, Washington, DC., USA
| | - William Mobley
- Department of Neurosciences, University of California, San Diego, CA, USA
| | - Paul E. Moore
- Division of Allergy, Immunology, and Pulmonology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Nicolas M. Oreskovic
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA; Department of Internal Medicine, Massachusetts General Hospital, Boston, Mass
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ricardo S. Osorio
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - David Patterson
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
- Eleanor Roosevelt Institute, University of Denver, Denver, CO, USA; Department of Biological Sciences, University of Denver, Denver, CO, USA; Molecular and Cellular Biophysics Program, University of Denver, Denver, CO, USA
| | - Sonja A. Rasmussen
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL; Department of Epidemiology, University of Florida College of Public Health and Health Professions and College of Medicine, Gainesville, FL
| | - Roger H. Reeves
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nancy Roizen
- Department of Pediatrics, UH/Rainbow Babies and Children’s Hospital and Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Stephanie Santoro
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Down Syndrome Program, Division of Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| | - Stephanie L. Sherman
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Nasreen Talib
- Division of General Pediatrics, Children’s Mercy Kansas City, 2401 Gillham Road, Kansas City, MO, USA
| | - Ignacio E. Tapia
- Sleep Center, Division of Pulmonary Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kyle M. Walsh
- Division of Neuro-epidemiology, Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Steven F. Warren
- Institute for Life Span Studies, University of Kansas, Lawrence, KS, USA
| | - A. Nicole White
- Research Foundation, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Guang William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John S. Yi
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
16
|
RCAN1.4 mediates high glucose-induced matrix production by stimulating mitochondrial fission in mesangial cells. Biosci Rep 2021; 40:221739. [PMID: 31894838 PMCID: PMC6970086 DOI: 10.1042/bsr20192759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/30/2019] [Accepted: 12/18/2019] [Indexed: 11/17/2022] Open
Abstract
High glucose (HG)-induced mitochondrial dynamic changes and oxidative damage are closely related to the development and progression of diabetic kidney disease (DKD). Recent studies suggest that regulators of calcineurin 1 (RCAN1) is involved in the regulation of mitochondrial function in different cell types, so we investigate the role of RCAN1 in mitochondrial dynamics under HG ambience in rat glomerular mesangial cells (MCs). MCs subjected to HG exhibited an isoform-specific up-regulation of RCAN1.4 at both mRNA and protein levels. RCAN1.4 overexpression induced translocation of Dynamin related protein 1 (Drp1) to mitochondria, mitochondrial fragmentation and depolarization, accompanied by increased matrix production under normal glucose and HG ambience. In contrast, decreasing the expression of RCAN1.4 by siRNA inhibited HG-induced mitochondrial fragmentation and matrix protein up-regulation. Moreover, both mitochondrial fission inhibitor Mdivi-1 and Drp1 shRNA prevented RCAN1.4-induced fibronectin up-regulation, suggesting that RCAN1.4-induced matrix production is dependent on its modulation of mitochondrial fission. Although HG-induced RCAN1.4 up-regulation was achieved by activating calcineurin, RCAN1.4-mediated mitochondrial fragmentation and matrix production is independent of calcineurin activity. These results provide the first evidence for the HG-induced RCAN1.4 up-regulation involving increased mitochondrial fragmentation, leading to matrix protein up-regulation.
Collapse
|
17
|
Lanzillotta C, Tramutola A, Di Giacomo G, Marini F, Butterfield DA, Di Domenico F, Perluigi M, Barone E. Insulin resistance, oxidative stress and mitochondrial defects in Ts65dn mice brain: A harmful synergistic path in down syndrome. Free Radic Biol Med 2021; 165:152-170. [PMID: 33516914 DOI: 10.1016/j.freeradbiomed.2021.01.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/12/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023]
Abstract
Dysregulation of brain insulin signaling with reduced downstream neuronal survival and plasticity mechanisms are fundamental abnormalities observed in Alzheimer disease (AD). This phenomenon, known as brain insulin resistance, is associated with poor cognitive performance and is driven by the inhibition of IRS1. Since Down syndrome (DS) and AD neuropathology share many common features, we investigated metabolic aspects of neurodegeneration in DS and whether they contribute to early onset AD in DS. We evaluated levels and activation of proteins belonging to the insulin signaling pathway (IR, IRS1, BVR-A, MAPK, PTEN, Akt, GSK3β, PKCζ, AS160, GLUT4) in the frontal cortex of Ts65dn (DS model) (n = 5-6/group) and euploid mice (n = 6/group) at different ages (1, 3, 9 and 18 months). Furthermore, we analyzed whether changes of brain insulin signaling were associated with alterations of: (i) proteins regulating brain energy metabolism (mitochondrial complexes, hexokinase-II, Sirt1); (ii) oxidative stress (OS) markers (iii) APP cleavage; and (iv) proteins mediating synaptic plasticity mechanisms (PSD95, syntaxin-1 and BDNF). Ts65dn mice showed an overall impairment of the above-mentioned pathways, mainly characterized by defects of proteins activation state. Such alterations start early in life (at 1 month, during brain maturation). In particular, accumulation of inhibited IRS1, together with the uncoupling among the proteins downstream from IRS1 (brain insulin resistance), characterize Ts65dn mice. Furthermore, reduced levels of mitochondrial complexes and Sirt1, as well as increased indices of OS also were observed. These alterations precede the accumulation of APP-C99 in Ts65dn mice. Tellingly, oxidative stress levels were negatively associated with IR, IRS1 and AS160 activation as well as mitochondrial complexes levels in Ts65dn mice, suggesting a role for oxidative stress in the observed alterations. We propose that a close link exists among brain insulin resistance, mitochondrial defects and OS that contributes to brain dysfunctions observed in DS, likely favoring the development of AD in DS.
Collapse
Affiliation(s)
- Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Graziella Di Giacomo
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Federico Marini
- Department of Chemistry, Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40506-0055, USA
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy.
| |
Collapse
|
18
|
Moreau M, Benhaddou S, Dard R, Tolu S, Hamzé R, Vialard F, Movassat J, Janel N. Metabolic Diseases and Down Syndrome: How Are They Linked Together? Biomedicines 2021; 9:biomedicines9020221. [PMID: 33671490 PMCID: PMC7926648 DOI: 10.3390/biomedicines9020221] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
Down syndrome is a genetic disorder caused by the presence of a third copy of chromosome 21, associated with intellectual disabilities. Down syndrome is associated with anomalies of both the nervous and endocrine systems. Over the past decades, dramatic advances in Down syndrome research and treatment have helped to extend the life expectancy of these patients. Improved life expectancy is obviously a positive outcome, but it is accompanied with the need to address previously overlooked complications and comorbidities of Down syndrome, including obesity and diabetes, in order to improve the quality of life of Down syndrome patients. In this focused review, we describe the associations between Down syndrome and comorbidities, obesity and diabetes, and we discuss the understanding of proposed mechanisms for the association of Down syndrome with metabolic disorders. Drawing molecular mechanisms through which Type 1 diabetes and Type 2 diabetes could be linked to Down syndrome could allow identification of novel drug targets and provide therapeutic solutions to limit the development of metabolic and cognitive disorders.
Collapse
Affiliation(s)
- Manon Moreau
- Laboratoire Processus Dégénératifs, Université de Paris, BFA, UMR 8251, CNRS, Stress et Vieillissemen, F-75013 Paris, France; (M.M.); (S.B.); (R.D.)
| | - Soukaina Benhaddou
- Laboratoire Processus Dégénératifs, Université de Paris, BFA, UMR 8251, CNRS, Stress et Vieillissemen, F-75013 Paris, France; (M.M.); (S.B.); (R.D.)
| | - Rodolphe Dard
- Laboratoire Processus Dégénératifs, Université de Paris, BFA, UMR 8251, CNRS, Stress et Vieillissemen, F-75013 Paris, France; (M.M.); (S.B.); (R.D.)
- Genetics Deptartment, CHI Poissy St Germain-en-Laye, F-78300 Poissy, France;
- Université Paris-Saclay, UVSQ, INRAE, ENVA, BREED, F-78350 Jouy-en-Josas, France
| | - Stefania Tolu
- Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France; (S.T.); (R.H.); (J.M.)
| | - Rim Hamzé
- Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France; (S.T.); (R.H.); (J.M.)
| | - François Vialard
- Genetics Deptartment, CHI Poissy St Germain-en-Laye, F-78300 Poissy, France;
- Université Paris-Saclay, UVSQ, INRAE, ENVA, BREED, F-78350 Jouy-en-Josas, France
| | - Jamileh Movassat
- Laboratoire de Biologie et Pathologie du Pancréas Endocrine, Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France; (S.T.); (R.H.); (J.M.)
| | - Nathalie Janel
- Laboratoire Processus Dégénératifs, Université de Paris, BFA, UMR 8251, CNRS, Stress et Vieillissemen, F-75013 Paris, France; (M.M.); (S.B.); (R.D.)
- Correspondence: ; Tel.: +33-1-57-27-83-60; Fax: +33-1-57-27-83-54
| |
Collapse
|
19
|
Sang XY, Xiao JJ, Liu Q, Zhu R, Dai JJ, Zhang C, Yu H, Yang SJ, Zhang BF. Regulators of calcineurin 1 deficiency attenuates tubulointerstitial fibrosis through improving mitochondrial fitness. FASEB J 2020; 34. [PMID: 32896034 DOI: 10.1096/fj.202000781rrr] [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] [Received: 04/01/2020] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023]
Abstract
Renal fibrosis is the common pathological process of various chronic kidney diseases (CKD). Recent studies indicate that mitochondrial fragmentation is closely associated with renal fibrosis in CKD. However, the molecular mechanisms leading to mitochondrial fragmentation remain to be elucidated. The present study investigated the role of regulators of calcineurin 1 (RCAN1) in mitochondrial fission and renal interstitial fibrosis using conditional knockout mice in which RCAN1 was genetically deleted in tubular epithelial cells (TECs). TEC-specific deletion of RCAN1 attenuated tubulointerstitial fibrosis and epithelial to mesenchymal transition (EMT)-like phenotype change after unilateral ureteral obstruction (UUO) and ischemia reperfusion injury (IRI) through suppressing TGF-β1/Smad3 signaling pathway. TEC-specific deletion of RCAN1 also reduced the tubular apoptosis after UUO by inhibiting cytochrome c/caspase-9 pathway. Ultrastructure analysis revealed a marked decrease in mitochondrial fragmentation in TECs of RCAN1-deficient mice in experimental CKD models. The expression of mitochondrial profission proteins dynamin-related protein 1 (Drp1) and mitochondrial fission factor (Mff) was also downregulated in obstructed kidney of TEC-specific RCAN1-deficient mice. Furthermore, TEC-specific deletion of RCAN1 attenuated the dysfunctional tubular autophagy by regulating PINK1/Parkin-induced mitophagy in CKD. RCAN1 knockdown and knockout similarly improved the mitochondrial quality control in HK-2 cells and primary cultured mouse tubular cells stimulated by TGF-β1. Put together, our data indicated that RCAN1 plays an important role in the progression of tubulointerstitial fibrosis through regulating the mitochondrial quality. Therefore, targeting RCAN1 may provide a potential therapeutic approach in CKD.
Collapse
Affiliation(s)
- Xue-Yu Sang
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Jing-Jie Xiao
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Qing Liu
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Rui Zhu
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Jia-Jia Dai
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Cheng Zhang
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Hong Yu
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Si-Jun Yang
- ABSL-3 Laboratory at the Center for Animal Experiment, State Key Laboratory of Virology, Wuhan University School of Medicine, Wuhan, P.R. China
| | - Bai-Fang Zhang
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| |
Collapse
|
20
|
Lee SK, Ahnn J. Regulator of Calcineurin (RCAN): Beyond Down Syndrome Critical Region. Mol Cells 2020; 43:671-685. [PMID: 32576715 PMCID: PMC7468584 DOI: 10.14348/molcells.2020.0060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/23/2020] [Accepted: 05/25/2020] [Indexed: 12/19/2022] Open
Abstract
The regulator of calcineurin (RCAN) was first reported as a novel gene called DSCR1, encoded in a region termed the Down syndrome critical region (DSCR) of human chromosome 21. Genome sequence comparisons across species using bioinformatics revealed three members of the RCAN gene family, RCAN1, RCAN2, and RCAN3, present in most jawed vertebrates, with one member observed in most invertebrates and fungi. RCAN is most highly expressed in brain and striated muscles, but expression has been reported in many other tissues, as well, including the heart and kidneys. Expression levels of RCAN homologs are responsive to external stressors such as reactive oxygen species, Ca2+, amyloid β, and hormonal changes and upregulated in pathological conditions, including Alzheimer's disease, cardiac hypertrophy, diabetes, and degenerative neuropathy. RCAN binding to calcineurin, a Ca2+/calmodulin-dependent phosphatase, inhibits calcineurin activity, thereby regulating different physiological events via dephosphorylation of important substrates. Novel functions of RCANs have recently emerged, indicating involvement in mitochondria homeostasis, RNA binding, circadian rhythms, obesity, and thermogenesis, some of which are calcineurin-independent. These developments suggest that besides significant contributions to DS pathologies and calcineurin regulation, RCAN is an important participant across physiological systems, suggesting it as a favorable therapeutic target.
Collapse
Affiliation(s)
- Sun-Kyung Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Research Institute for Natural Sciences, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Joohong Ahnn
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
- Research Institute for Natural Sciences, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
21
|
Muramatsu M, Nakagawa S, Osawa T, Toyono T, Uemura A, Kidoya H, Takakura N, Usui T, Ryeom S, Minami T. Loss of Down Syndrome Critical Region-1 Mediated-Hypercholesterolemia Accelerates Corneal Opacity Via Pathological Neovessel Formation. Arterioscler Thromb Vasc Biol 2020; 40:2425-2439. [PMID: 32787520 DOI: 10.1161/atvbaha.120.315003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE The calcineurin-NFAT (nuclear factor for activated T cells)-DSCR (Down syndrome critical region)-1 pathway plays a crucial role as the downstream effector of VEGF (vascular endothelial growth factor)-mediated tumor angiogenesis in endothelial cells. A role for DSCR-1 in different organ microenvironment such as the cornea and its role in ocular diseases is not well understood. Corneal changes can be indicators of various disease states and are easily detected through ocular examinations. Approach and Results: The presentation of a corneal arcus or a corneal opacity due to lipid deposition in the cornea often indicates hyperlipidemia and in most cases, hypercholesterolemia. Although the loss of Apo (apolipoprotein) E has been well characterized and is known to lead to elevated serum cholesterol levels, there are few corneal changes observed in ApoE-/- mice. In this study, we show that the combined loss of ApoE and DSCR-1 leads to a dramatic increase in serum cholesterol levels and severe corneal opacity with complete penetrance. The cornea is normally maintained in an avascular state; however, loss of Dscr-1 is sufficient to induce hyper-inflammatory and -oxidative condition, increased corneal neovascularization, and lymphangiogenesis. Furthermore, immunohistological analysis and genome-wide screening revealed that loss of Dscr-1 in mice triggers increased immune cell infiltration and upregulation of SDF (stromal derived factor)-1 and its receptor, CXCR4 (C-X-C motif chemokine ligand receptor-4), potentiating this signaling axis in the cornea, thereby contributing to pathological corneal angiogenesis and opacity. CONCLUSIONS This study is the first demonstration of the critical role for the endogenous inhibitor of calcineurin, DSCR-1, and pathological corneal angiogenesis in hypercholesterolemia induced corneal opacity.
Collapse
Affiliation(s)
- Masashi Muramatsu
- Division of Molecular and Vascular Biology, IRDA, Kumamoto University, Japan (M.M., T.M.)
| | - Suguru Nakagawa
- Division of Genome Science (S.N.), RCAST, the University of Tokyo, Japan.,Department Ophthalmology, Graduate School of Medicine, the University of Tokyo, Japan (S.N., T.T., T.U.)
| | - Tsuyoshi Osawa
- Integrative Nutriomics (T.O.), RCAST, the University of Tokyo, Japan
| | - Tetsuya Toyono
- Department Ophthalmology, Graduate School of Medicine, the University of Tokyo, Japan (S.N., T.T., T.U.)
| | - Akiyoshi Uemura
- Department Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, Japan (A.U.)
| | - Hiroyasu Kidoya
- Department Signal Transduction, RIMD, Osaka University, Japan (H.K., N.T.)
| | - Nobuyuki Takakura
- Department Signal Transduction, RIMD, Osaka University, Japan (H.K., N.T.)
| | - Tomohiko Usui
- Department Ophthalmology, Graduate School of Medicine, the University of Tokyo, Japan (S.N., T.T., T.U.)
| | - Sandra Ryeom
- Department Cancer Biology, University of Pennsylvania (S.R.)
| | - Takashi Minami
- Division of Molecular and Vascular Biology, IRDA, Kumamoto University, Japan (M.M., T.M.)
| |
Collapse
|
22
|
Dierssen M, Fructuoso M, Martínez de Lagrán M, Perluigi M, Barone E. Down Syndrome Is a Metabolic Disease: Altered Insulin Signaling Mediates Peripheral and Brain Dysfunctions. Front Neurosci 2020; 14:670. [PMID: 32733190 PMCID: PMC7360727 DOI: 10.3389/fnins.2020.00670] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/02/2020] [Indexed: 12/19/2022] Open
Abstract
Down syndrome (DS) is the most frequent chromosomal abnormality that causes intellectual disability, resulting from the presence of an extra complete or segment of chromosome 21 (HSA21). In addition, trisomy of HSA21 contributes to altered energy metabolism that appears to be a strong determinant in the development of pathological phenotypes associated with DS. Alterations include, among others, mitochondrial defects, increased oxidative stress levels, impaired glucose, and lipid metabolism, finally resulting in reduced energy production and cellular dysfunctions. These molecular defects seem to account for a high incidence of metabolic disorders, i.e., diabetes and/or obesity, as well as a higher risk of developing Alzheimer’s disease (AD) in DS. A dysregulation of the insulin signaling with reduced downstream pathways represents a common pathophysiological aspect in the development of both peripheral and central alterations leading to diabetes/obesity and AD. This is further strengthened by evidence showing that the molecular mechanisms responsible for such alterations appear to be similar between peripheral organs and brain. Considering that DS subjects are at high risk to develop either peripheral or brain metabolic defects, this review will discuss current knowledge about the link between trisomy of HSA21 and defects of insulin and insulin-related pathways in DS. Drawing the molecular signature underlying these processes in DS is a key challenge to identify novel drug targets and set up new prevention strategies aimed to reduce the impact of metabolic disorders and cognitive decline.
Collapse
Affiliation(s)
- Mara Dierssen
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain.,Human Pharmacology and Clinical Neurosciences Research Group, Neurosciences Research Program, Hospital Del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Marta Fructuoso
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - María Martínez de Lagrán
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
23
|
Moriwaki T, Abe S, Oshimura M, Kazuki Y. Transchromosomic technology for genomically humanized animals. Exp Cell Res 2020; 390:111914. [PMID: 32142854 DOI: 10.1016/j.yexcr.2020.111914] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/16/2020] [Accepted: 02/19/2020] [Indexed: 12/15/2022]
Abstract
"Genomically" humanized animals are invaluable tools for generating human disease models and for biomedical research. Humanized animal models have generally been developed via conventional transgenic technologies; however, conventional gene delivery vectors such as viruses, plasmids, bacterial artificial chromosomes, P1 phase-derived artificial chromosomes, and yeast artificial chromosomes have limitations for transgenic animal creation as their loading gene capacity is restricted, and the expression of transgenes is unstable. Transchromosomic (Tc) techniques using mammalian artificial chromosomes, including human chromosome fragments, human artificial chromosomes, and mouse artificial chromosomes, have overcome these limitations. These tools can carry multiple genes or Mb-sized genomic loci and their associated regulatory elements, which has facilitated the creation of more useful and complex transgenic models for human disease, drug development, and humanized animal research. This review describes the history of Tc animal development, the applications of Tc animals, and future prospects.
Collapse
Affiliation(s)
- Takashi Moriwaki
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| | - Satoshi Abe
- Trans Chromosomics, Inc., 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| | - Mitsuo Oshimura
- Trans Chromosomics, Inc., 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan; Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| | - Yasuhiro Kazuki
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan; Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan.
| |
Collapse
|
24
|
Li X, Chan LWC, Li X, Liu C, Yang G, Gao J, Dai M, Wang Y, Xie Z, Liu J, Zhou F, Zheng T, Feng D, Guo S, Li H, Sun K, Yang S. Obesity-Induced Regulator of Calcineurin 1 Overexpression Leads to β-Cell Failure Through Mitophagy Pathway Inhibition. Antioxid Redox Signal 2020; 32:413-428. [PMID: 31822118 DOI: 10.1089/ars.2019.7806] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Aims: Type 2 diabetes (T2D) is associated with pancreatic β-cell dysfunction, manifested by reduced glucose-stimulated insulin secretion (GSIS). The regulator of calcineurin 1 (RCAN1) in islets is an endogenous inhibitor of calcium-activated protein phosphatase. Previous studies have indicated that global RCAN1 overexpression under high nutrient stress is involved in insulin resistance in T2D. However, the specific role and mechanism of this gene's overexpression in pancreatic β-cells have not been thoroughly elucidated to date. Results: In this study, we showed that mice overexpressing islet-specific RCAN1 exhibited a prediabetic phenotype with markedly reduced GSIS under nutrient stress. Overexpression of RCAN1 increased the autophagy-associated DNA methylation level of Beclin-1 suppressing the induction of autophagy, affected the protein kinase B, and downregulated the activation of mammalian target of rapamycin, leading to Miro1-mediated mitophagy deficiency. Furthermore, the exacerbated impairment of autophagy induction and mitophagy flux failures induced β-cell apoptosis, resulting in GSIS impairment, lipid imbalance, and NOD-like receptor 3 proinflammation under high nutrient stress in mice. Innovation: Our present data identify a detrimental effect of RCAN1 overexpression on Miro1-mediated mitophagy deficiency and β-cell dysfunction in high-fat diet-fed RCAN1 overexpressing mice. Conclusion: Our results revealed that strategies targeting RCAN1 in vivo may provide a therapeutic target to enhance β-cell mitophagy quality and may determine the crucial factor in T2D development.
Collapse
Affiliation(s)
- Xujun Li
- ABSL-3 Laboratory at the Center for Animal Experiment, Institute of Animal Model for Human Disease, Wuhan University School of Medicine, Wuhan, People's Republic of China
| | - Lawrence W C Chan
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hong Kong, Hong Kong
| | - Xianyu Li
- Department of Pathophysiology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, People's Republic of China
| | - Chunyan Liu
- ABSL-3 Laboratory at the Center for Animal Experiment, Institute of Animal Model for Human Disease, Wuhan University School of Medicine, Wuhan, People's Republic of China
| | - Guohua Yang
- Demonstration Center for Experimental Basic Medicine Education, School of Basic Medical Science, Wuhan University, Wuhan, People's Republic of China
| | - Jianfeng Gao
- ABSL-3 Laboratory at the Center for Animal Experiment, Institute of Animal Model for Human Disease, Wuhan University School of Medicine, Wuhan, People's Republic of China
| | - Ming Dai
- ABSL-3 Laboratory at the Center for Animal Experiment, Institute of Animal Model for Human Disease, Wuhan University School of Medicine, Wuhan, People's Republic of China
| | - Yunxin Wang
- School of Medical Technology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Zhiwen Xie
- School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Junli Liu
- Shanghai Diabetes Research Institute, Shanghai JiaoTong University Affiliated 6th People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Tian Zheng
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Du Feng
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Shaodong Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
| | - Haojie Li
- Shanghai Center for Plant Stress Biology, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Kai Sun
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Sijun Yang
- ABSL-3 Laboratory at the Center for Animal Experiment, Institute of Animal Model for Human Disease, Wuhan University School of Medicine, Wuhan, People's Republic of China
| |
Collapse
|
25
|
Gutierrez-Hervas A, Gómez-Martínez S, Izquierdo-Gómez R, Veiga OL, Perez-Bey A, Castro-Piñero J, Marcos A. Inflammation and fatness in adolescents with and without Down syndrome: UP & DOWN study. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2020; 64:170-179. [PMID: 31858639 DOI: 10.1111/jir.12697] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND The main objective of this study was to describe the inflammatory status of adolescents with Down syndrome (DS) and their relationship with adiposity. METHODS Ninety-five adolescents with DS (44.2% girls) and a control group of 113 adolescents (47.8% girls), aged between 11 and 18 years old, from the UP & DOWN study were included in this substudy. Serum C-reactive protein, C3 and C4 complement factors, total proteins, interleukin-6, tumour necrosis factor-α, insulin, cortisol, leptin, adiponectin, galactin-3 and visfatin were analysed; homeostatic model assessment index was calculated. In order to evaluate adiposity, we measured the following body fat variables: weight, height, waist circumference and skinfold thicknesses. Birth weight was obtained by questionnaire. In addition, body mass index, waist-to-height ratio (WHtR) and body fat percentage (BF%) were calculated. RESULTS Down syndrome group showed higher levels of body mass index, WHtR, waist circumference, BF% and lower birth weight than controls (P < 0.001). In the general linear model in the total sample, WHtR was positively associated with C3 and C4 (P < 0.001) as well as with leptin levels (P = 0.015). BF% was positively associated with total proteins (P = 0.093) and leptin levels (P < 0.001). DS was positively associated with total proteins (P < 0.001), C3 (P = 0.047) and C4 (P = 0.019). Despite the higher levels of adiposity found in DS group, no direct association was found between BF% and leptin levels, comparing with the control group. CONCLUSIONS These findings suggest that abdominal obesity should be controlled in adolescents because of its relationship with acute phase-inflammatory biomarkers but especially in DS adolescents who may show a peculiar metabolic status according to their relationship between adiposity and inflammatory biomarkers.
Collapse
Affiliation(s)
- A Gutierrez-Hervas
- Nursing Department, University of Alicante, Alicante, Spain
- Department of Nutrition and Metabolism (DMN), Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), Madrid, Spain
| | - S Gómez-Martínez
- Department of Nutrition and Metabolism (DMN), Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), Madrid, Spain
| | - R Izquierdo-Gómez
- Department of Physical Education, Faculty of Education Sciences, University of Cádiz, Puerto Real, Spain
- Department of Physical Education, Faculty of Education, Universidad Central de Chile, Santiago, Chile
| | - O L Veiga
- Department of Physical Education, Sport and Human Movement, Faculty of Education, Autonomous University of Madrid, Ciudad Universitaria de Cantoblanco, Madrid, Spain
| | - A Perez-Bey
- Department of Physical Education, Faculty of Education Sciences, University of Cádiz, Puerto Real, Spain
| | - J Castro-Piñero
- Department of Physical Education, Faculty of Education Sciences, University of Cádiz, Puerto Real, Spain
| | - A Marcos
- Department of Nutrition and Metabolism (DMN), Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), Madrid, Spain
| |
Collapse
|
26
|
Gross TJ, Doran E, Cheema AK, Head E, Lott IT, Mapstone M. Plasma metabolites related to cellular energy metabolism are altered in adults with Down syndrome and Alzheimer's disease. Dev Neurobiol 2019; 79:622-638. [PMID: 31419370 DOI: 10.1002/dneu.22716] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/02/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022]
Abstract
Down syndrome (DS) is a well-known neurodevelopmental disorder most commonly caused by trisomy of chromosome 21. Because individuals with DS almost universally develop heavy amyloid burden and Alzheimer's disease (AD), biomarker discovery in this population may be extremely fruitful. Moreover, any AD biomarker in DS that does not directly involve amyloid pathology may be of high value for understanding broader mechanisms of AD generalizable to the neurotypical population. In this retrospective biomarker discovery study, we examined banked peripheral plasma samples from 78 individuals with DS who met clinical criteria for AD at the time of the blood draw (DS-AD) and 68 individuals with DS who did not (DS-NAD). We measured the relative abundance of approximately 5,000 putative features in the plasma using untargeted mass spectrometry (MS). We found significantly higher levels of a peak putatively annotated as lactic acid in the DS-AD group (q = .014), a finding confirmed using targeted MS (q = .011). Because lactate is the terminal product of glycolysis and subsequent lactic acid fermentation, we performed additional targeted MS focusing on central carbon metabolism which revealed significantly increased levels of pyruvic (q = .03) and methyladipic (q = .03) acids in addition to significantly lower levels of uridine (q = .007) in the DS-AD group. These data suggest that AD in DS is accompanied by a shift from aerobic respiration toward the less efficient fermentative metabolism and that bioenergetically derived metabolites observable in peripheral blood may be useful for detecting this shift.
Collapse
Affiliation(s)
- Thomas J Gross
- Department of Neurology, The University of California, Irvine, Irvine, California
| | - Eric Doran
- Department of Pediatrics, The University of California, Irvine, Irvine, California
| | - Amrita K Cheema
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, The University of California, Irvine, Irvine, California
| | - Ira T Lott
- Department of Pediatrics, The University of California, Irvine, Irvine, California
| | - Mark Mapstone
- Department of Neurology, The University of California, Irvine, Irvine, California
| |
Collapse
|
27
|
Magge SN, Zemel BS, Pipan ME, Gidding SS, Kelly A. Cardiometabolic Risk and Body Composition in Youth With Down Syndrome. Pediatrics 2019; 144:peds.2019-0137. [PMID: 31315916 PMCID: PMC6855833 DOI: 10.1542/peds.2019-0137] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/17/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Whether BMI captures adiposity and cardiometabolic risk in Down syndrome (DS), a condition associated with obesity, short stature, and altered body proportions, is not known. We compared cardiometabolic risk measures in youth with DS and typically developing matched controls. METHODS Youth with (n = 150) and without (n = 103) DS of comparable age (10-20 years), sex, race, ethnicity, and BMI percentile underwent whole-body dual-energy X-ray absorptiometry, fasting glucose, insulin, lipids, lipoprotein particles, inflammatory factors, and when BMI percentile ≥85, an oral glucose tolerance test. RESULTS Sixty-four percent of youth with DS had BMI percentile ≥85. Among these, no difference in glucose, insulin, or insulin resistance was detected, but prediabetes was more prevalent with DS (26.4% vs 10.3%; P = .025) after adjustment for demographics, pubertal status, and BMI z score (odds ratio = 3.2; P = .026). Among all participants, those with DS had higher low-density lipoprotein cholesterol (median 107 [interquartile range 89-128] vs 88.5 [79-103] mg/dL; P < .00005), triglycerides (89.5 [73-133] vs 71.5 [56-104] mg/dL; P < .00005), non-high-density lipoprotein cholesterol (non-HDL-C; 128 [104-153] vs 107 [92-123] mg/dL; P < .00005), and triglycerides/HDL-C (2.2 [1.6-3.4] vs 1.7 [1.1-2.5] mg/dL; P = .0003) and lower levels of HDL-C (41 [36.5-47] vs 45 [37-53] mg/dL; P = .012). DS youth had higher high-sensitivity C-reactive protein, interleukin-6, small low-density lipoprotein particles (LDL-P), and total LDL-P, but similar LDL-P size. Youth with DS had less visceral fat (VFAT), fat mass, and lean mass for BMI z score, but greater VFAT at higher fat mass. However, VFAT did not fully explain the increased prevalence of dyslipidemia or prediabetes in youth with DS. CONCLUSIONS Despite similar insulin resistance, youth with DS had greater prevalence of dyslipidemia and prediabetes than typically developing youth, which was not fully explained by VFAT.
Collapse
Affiliation(s)
- Sheela N. Magge
- Division of Endocrinology and Diabetes, School of Medicine and Health Sciences, The George Washington University and Clinical and Translational Science Institute at Children’s National, Children’s Research Institute, Children’s National Health System, Washington, District of Columbia; Divisions of
| | - Babette S. Zemel
- Gastroenterology, Hepatology, and Nutrition,,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania; and
| | - Mary E. Pipan
- Developmental Behavioral Pediatrics, and,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania; and
| | | | - Andrea Kelly
- Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania;,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania; and
| |
Collapse
|
28
|
Delikkaya B, Moriel N, Tong M, Gallucci G, de la Monte SM. Altered expression of insulin-degrading enzyme and regulator of calcineurin in the rat intracerebral streptozotocin model and human apolipoprotein E-ε4-associated Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2019; 11:392-404. [PMID: 31193223 PMCID: PMC6522644 DOI: 10.1016/j.dadm.2019.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION This study assesses insulin-degrading enzyme (IDE) and regulator of calcineurin 1 (RCAN1) as potential mediators of brain insulin deficiency and neurodegeneration in experimental and human Alzheimer's disease (AD). METHODS Temporal lobes from Long Evans rats treated with intracerebral streptozotocin or vehicle and postmortem frontal lobes from humans with normal aging AD (Braak 0-2), moderate (Braak 3-4) AD, or advanced (Braak 5-6) AD were used to measure IDE and RCAN mRNA and protein. RESULTS Intracerebral streptozotocin significantly increased IDE and RCAN mRNA and protein. In humans with apolipoprotein E (ApoE) ε3/ε4 or ε4/ε4 and AD, IDE was elevated at Braak 3-4, but at Braak 5-6, IDE expression was significantly reduced. RCAN1 mRNA was similarly reduced in ApoE ε4+ patients with moderate or severe AD, whereas RCAN1 protein declined with the severity of AD and ApoE ε4 dose. DISCUSSION The findings suggest that IDE and RCAN1 differentially modulate brain insulin signaling in relation to AD severity and ApoE genotype.
Collapse
Affiliation(s)
- Büşra Delikkaya
- Istanbul University-Cerrahpasa Cerrahpasa Medical Faculty, Istanbul, Turkey
| | - Natalia Moriel
- Department of Medicine, Rhode Island Hospital, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Providence, RI, USA,Alpert Medical School of Brown University, Providence, RI, USA
| | - Gina Gallucci
- Department of Medicine, Rhode Island Hospital, Providence, RI, USA
| | - Suzanne M. de la Monte
- Department of Medicine, Rhode Island Hospital, Providence, RI, USA,Alpert Medical School of Brown University, Providence, RI, USA,Departments of Neurology and Neurosurgery, Rhode Island Hospital, Providence, RI, USA,Department of Pathology and Laboratory Medicine, Providence VA Medical Center, Providence, RI, USA,Corresponding author. Tel.: +401-444-7364; Fax: +401-444-2939.
| |
Collapse
|
29
|
Keating D. Regulator of Calcineurin 1 (RCAN1) helps coordinate whole body metabolism and limit energy expenditure. Obes Res Clin Pract 2019. [DOI: 10.1016/j.orcp.2018.11.079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
30
|
de la Monte SM, Tong M, Daiello LA, Ott BR. Early-Stage Alzheimer's Disease Is Associated with Simultaneous Systemic and Central Nervous System Dysregulation of Insulin-Linked Metabolic Pathways. J Alzheimers Dis 2019; 68:657-668. [PMID: 30775986 PMCID: PMC10084886 DOI: 10.3233/jad-180906] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Brain insulin resistance is a well-recognized abnormality in Alzheimer's disease (AD) and the likely mediator of impaired glucose utilization that emerges early and progresses with disease severity. Moreover, the rates of mild cognitive impairment (MCI) or AD are significantly greater in people with diabetes mellitus or obesity. OBJECTIVE This study was designed to determine whether systemic and central nervous system (CNS) insulin resistant disease states emerge together and thus may be integrally related. METHODS Insulin-related molecules were measured in paired human serum and cerebrospinal fluid (CSF) samples from 19 with MCI or early AD, and 21 controls using a multiplex ELISA platform. RESULTS In MCI/AD, both the CSF and serum samples had significantly elevated mean levels of C-peptide and an incretin, and reduced expression of Visfatin, whereas only CSF showed significant reductions in insulin and leptin and only serum had increased glucagon, PAI-1, and ghrelin. Although the overall CSF and serum responses reflected insulin resistance together with insulin deficiency, the specific alterations measured in CSF and serum were different. CONCLUSION In MCI and early-stage AD, CNS and systemic insulin-related metabolic dysfunctions, including insulin resistance, occur simultaneously, suggesting that they are integrally related and possibly mediated similar pathogenic factors.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Pathology and Laboratory Medicine (Neuropathology), Rhode Island Hospital, the Providence VA Medical Center, and the Alpert Medical School of Brown University, Providence, RI, USA.,Department of Neurology, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA.,Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Lori A Daiello
- Department of Neurology, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA.,The Alzheimer's Disease and Memory Disorders Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Brian R Ott
- Department of Neurology, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA.,The Alzheimer's Disease and Memory Disorders Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
31
|
A single extra copy of Down syndrome critical region 1-4 results in impaired hepatic glucose homeostasis. Mol Metab 2018; 21:82-89. [PMID: 30583978 PMCID: PMC6407364 DOI: 10.1016/j.molmet.2018.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/29/2018] [Accepted: 12/03/2018] [Indexed: 01/19/2023] Open
Abstract
Objectives During fasting, hepatic gluconeogenesis is induced to maintain energy homeostasis. Moreover, abnormal dysregulation of hepatic glucose production is commonly observed in type 2 diabetes. However, the signaling components controlling hepatic glucose production to maintain normal glucose levels are not fully understood. Here, we examined the physiological role of Down syndrome critical region 1–4 (DSCR1-4), an endogenous calcineurin signaling inhibitor in the liver that mediates metabolic adaptation to fasting. Methods We assessed the effect of cyclosporine A, an inhibitor of calcineurin signaling on gluconeogenic gene expression in primary hepatocytes. DSCR1-4 expression was examined in diet- and genetically-induced mouse models of obesity. We also investigated the metabolic phenotype of a single extra copy of DSCR1-4 in transgenic mice and how DSCR1-4 regulates glucose homeostasis in the liver. Results Treatment with cyclosporin A increased hepatic glucose production and gluconeogenic gene expression. The expression of DSCR1-4 was induced by refeeding and overexpressed in obese mouse livers. Moreover, transgenic mice with a single extra copy of DSCR1-4 exhibited pyruvate intolerance and impaired glucose homeostasis. Mechanistically, DSCR1-4 overexpression increased phosphorylation of the cAMP response element-binding protein, which led to elevated expression levels of gluconeogenic genes and, thus, enhanced hepatic glucose production during fasting. Conclusion A single extra copy of DSCR1-4 results in dysregulated hepatic glucose homeostasis and pyruvate intolerance. Our findings suggest that nutrient-sensitive DSCR1-4 is a novel target for controlling hepatic gluconeogenesis in diabetes. DSCR1 mRNA and protein levels are increased in livers upon nutrient availability. DSCR1-4 is overexpressed in diet- or genetically induced obesity. DSCR1-4 trisomy mice exhibit impaired glucose homeostasis and pyruvate intolerance. Trisomy of DSCR1-4 leads to increased hepatic glucose production.
Collapse
|
32
|
Rotter D, Peiris H, Grinsfelder DB, Martin AM, Burchfield J, Parra V, Hull C, Morales CR, Jessup CF, Matusica D, Parks BW, Lusis AJ, Nguyen NUN, Oh M, Iyoke I, Jakkampudi T, McMillan DR, Sadek HA, Watt MJ, Gupta RK, Pritchard MA, Keating DJ, Rothermel BA. Regulator of Calcineurin 1 helps coordinate whole-body metabolism and thermogenesis. EMBO Rep 2018; 19:embr.201744706. [PMID: 30389725 DOI: 10.15252/embr.201744706] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/12/2018] [Accepted: 10/05/2018] [Indexed: 12/12/2022] Open
Abstract
Increasing non-shivering thermogenesis (NST), which expends calories as heat rather than storing them as fat, is championed as an effective way to combat obesity and metabolic disease. Innate mechanisms constraining the capacity for NST present a fundamental limitation to this approach, yet are not well understood. Here, we provide evidence that Regulator of Calcineurin 1 (RCAN1), a feedback inhibitor of the calcium-activated protein phosphatase calcineurin (CN), acts to suppress two distinctly different mechanisms of non-shivering thermogenesis (NST): one involving the activation of UCP1 expression in white adipose tissue, the other mediated by sarcolipin (SLN) in skeletal muscle. UCP1 generates heat at the expense of reducing ATP production, whereas SLN increases ATP consumption to generate heat. Gene expression profiles demonstrate a high correlation between Rcan1 expression and metabolic syndrome. On an evolutionary timescale, in the context of limited food resources, systemic suppression of prolonged NST by RCAN1 might have been beneficial; however, in the face of caloric abundance, RCAN1-mediated suppression of these adaptive avenues of energy expenditure may now contribute to the growing epidemic of obesity.
Collapse
Affiliation(s)
- David Rotter
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Heshan Peiris
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, SA, Australia
| | - D Bennett Grinsfelder
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alyce M Martin
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, SA, Australia
| | - Jana Burchfield
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Valentina Parra
- Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Advanced Center for Chronic Diseases (ACCDiS) and Center for Exercise Metabolism and Cancer (CEMC), University of Chile, Santiago, Chile
| | - Christi Hull
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cyndi R Morales
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Claire F Jessup
- Department of Anatomy and Histology and Centre for Neuroscience, Flinders University, Adelaide, SA, Australia
| | - Dusan Matusica
- Department of Anatomy and Histology and Centre for Neuroscience, Flinders University, Adelaide, SA, Australia
| | - Brian W Parks
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Ngoc Uyen Nhi Nguyen
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Misook Oh
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Chemistry, Pohang University of Science and Technology, Pohang, South Korea
| | - Israel Iyoke
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tanvi Jakkampudi
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - D Randy McMillan
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Children's Medical Centre, Dallas, TX, USA
| | - Hesham A Sadek
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Matthew J Watt
- The Department of Physiology and Monash Biomedicine Discovery Institute, Metabolic Disease and Obesity Program, Monash University, Clayton, Vic., Australia
| | - Rana K Gupta
- Touchstone Diabetes Center and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Melanie A Pritchard
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Vic., Australia
| | - Damien J Keating
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, SA, Australia .,South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Beverly A Rothermel
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA .,Department of Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| |
Collapse
|
33
|
Herault Y, Delabar JM, Fisher EMC, Tybulewicz VLJ, Yu E, Brault V. Rodent models in Down syndrome research: impact and future opportunities. Dis Model Mech 2018; 10:1165-1186. [PMID: 28993310 PMCID: PMC5665454 DOI: 10.1242/dmm.029728] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Down syndrome is caused by trisomy of chromosome 21. To date, a multiplicity of mouse models with Down-syndrome-related features has been developed to understand this complex human chromosomal disorder. These mouse models have been important for determining genotype-phenotype relationships and identification of dosage-sensitive genes involved in the pathophysiology of the condition, and in exploring the impact of the additional chromosome on the whole genome. Mouse models of Down syndrome have also been used to test therapeutic strategies. Here, we provide an overview of research in the last 15 years dedicated to the development and application of rodent models for Down syndrome. We also speculate on possible and probable future directions of research in this fast-moving field. As our understanding of the syndrome improves and genome engineering technologies evolve, it is necessary to coordinate efforts to make all Down syndrome models available to the community, to test therapeutics in models that replicate the whole trisomy and design new animal models to promote further discovery of potential therapeutic targets. Summary: Mouse models have boosted therapeutic options for Down syndrome, and improved models are being developed to better understand the pathophysiology of this genetic condition.
Collapse
Affiliation(s)
- Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, 1 rue Laurent Fries, 67404 Illkirch, France .,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Université de Strasbourg, 67404 Illkirch, France.,T21 Research Society, Brain and Spine Institute (ICM), 75013 Paris
| | - Jean M Delabar
- T21 Research Society, Brain and Spine Institute (ICM), 75013 Paris.,Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, UMR8251, CNRS, 75205 Paris, France.,INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et la Moelle épinière, ICM, 75013 Paris, France.,Brain and Spine Institute (ICM) CNRS UMR7225, INSERM UMRS 975, 75013 Paris, France
| | - Elizabeth M C Fisher
- T21 Research Society, Brain and Spine Institute (ICM), 75013 Paris.,Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, WC1N 3BG, UK.,LonDownS Consortium, London, W1T 7NF UK
| | - Victor L J Tybulewicz
- T21 Research Society, Brain and Spine Institute (ICM), 75013 Paris.,LonDownS Consortium, London, W1T 7NF UK.,The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.,Department of Medicine, Imperial College, London, SW7 2AZ, UK
| | - Eugene Yu
- T21 Research Society, Brain and Spine Institute (ICM), 75013 Paris.,The Children's Guild Foundation Down Syndrome Research Program, Department of Cancer Genetics and Genetics Program, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.,Department of Cellular and Molecular Biology, Roswell Park Division of Graduate School, Genetics, Genomics and Bioinformatics Program, State University of New York at Buffalo, Buffalo, NY 14263, USA
| | - Veronique Brault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, 1 rue Laurent Fries, 67404 Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Université de Strasbourg, 67404 Illkirch, France
| |
Collapse
|
34
|
Coskun P, Helguera P, Nemati Z, Bohannan RC, Thomas J, Samuel SE, Argueta J, Doran E, Wallace DC, Lott IT, Busciglio J. Metabolic and Growth Rate Alterations in Lymphoblastic Cell Lines Discriminate Between Down Syndrome and Alzheimer's Disease. J Alzheimers Dis 2018; 55:737-748. [PMID: 27802222 DOI: 10.3233/jad-160278] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Deficits in mitochondrial function and oxidative stress play pivotal roles in Down syndrome (DS) and Alzheimer's disease (AD) and these alterations in mitochondria occur systemically in both conditions. OBJECTIVE We hypothesized that peripheral cells of elder subjects with DS exhibit disease-specific and dementia-specific metabolic features. To test this, we performed a comprehensive analysis of energy metabolism in lymphoblastic-cell-lines (LCLs) derived from subjects belonging to four groups: DS-with-dementia (DSAD), DS-without-dementia (DS), sporadic AD, and age-matched controls. METHODS LCLs were studied under regular or minimal feeding regimes with galactose or glucose as primary carbohydrate sources. We assessed metabolism under glycolysis or oxidative phosphorylation by quantifying cell viability, oxidative stress, ATP levels, mitochondrial membrane potential (MMP), mitochondrial calcium uptake, and autophagy. RESULTS DS and DSAD LCLs showed slower growth rates under minimal feeding. DS LCLs mainly dependent on mitochondrial respiration exhibited significantly slower growth and higher levels of oxidative stress compared to other groups. While ATP levels (under mitochondrial inhibitors) and mitochondrial calcium uptake were significantly reduced in DSAD and AD cells, MMP was decreased in DS, DSAD, and AD LCLs. Finally, DS LCLs showed markedly reduced levels of the autophagy marker LC3-II, underscoring the close association between metabolic dysfunction and impaired autophagy in DS. CONCLUSION There are significant mitochondrial functional changes in LCLs derived from DS, DSAD, and AD patients. Several parameters analyzed were consistently different between DS, DSAD, and AD lines suggesting that metabolic indicators between LCL groups may be utilized as biomarkers of disease progression and/or treatment outcomes.
Collapse
Affiliation(s)
- Pinar Coskun
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA
| | - Pablo Helguera
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA.,Instituto de Investigación Médica Mercedes y Martin Ferreyra, Córdoba, Argentina, USA
| | - Zahra Nemati
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA
| | - Ryan C Bohannan
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA
| | - Jean Thomas
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA
| | - Schriner E Samuel
- Department of Pharmaceutical Science, University of California, Irvine, CA, USA
| | - Jocelyn Argueta
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA
| | - Eric Doran
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine (CMEM), Children's Hospital of Philadelphia, and Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ira T Lott
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - Jorge Busciglio
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), and Center for the Neurobiology of Learning and Memory (CNLM), University of California, Irvine, CA, USA
| |
Collapse
|
35
|
Parra V, Altamirano F, Hernández-Fuentes CP, Tong D, Kyrychenko V, Rotter D, Pedrozo Z, Hill JA, Eisner V, Lavandero S, Schneider JW, Rothermel BA. Down Syndrome Critical Region 1 Gene, Rcan1, Helps Maintain a More Fused Mitochondrial Network. Circ Res 2018; 122:e20-e33. [PMID: 29362227 DOI: 10.1161/circresaha.117.311522] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 01/17/2018] [Accepted: 01/22/2018] [Indexed: 01/05/2023]
Abstract
RATIONALE The regulator of calcineurin 1 (RCAN1) inhibits CN (calcineurin), a Ca2+-activated protein phosphatase important in cardiac remodeling. In humans, RCAN1 is located on chromosome 21 in proximity to the Down syndrome critical region. The hearts and brains of Rcan1 KO mice are more susceptible to damage from ischemia/reperfusion (I/R); however, the underlying cause is not known. OBJECTIVE Mitochondria are key mediators of I/R damage. The goal of these studies was to determine the impact of RCAN1 on mitochondrial dynamics and function. METHODS AND RESULTS Using both neonatal and isolated adult cardiomyocytes, we show that, when RCAN1 is depleted, the mitochondrial network is more fragmented because of increased CN-dependent activation of the fission protein, DRP1 (dynamin-1-like). Mitochondria in RCAN1-depleted cardiomyocytes have reduced membrane potential, O2 consumption, and generation of reactive oxygen species, as well as a reduced capacity for mitochondrial Ca2+ uptake. RCAN1-depleted cardiomyocytes were more sensitive to I/R; however, pharmacological inhibition of CN, DRP1, or CAPN (calpains; Ca2+-activated proteases) restored protection, suggesting that in the absence of RCAN1, CAPN-mediated damage after I/R is greater because of a decrease in the capacity of mitochondria to buffer cytoplasmic Ca2+. Increasing RCAN1 levels by adenoviral infection was sufficient to enhance fusion and confer protection from I/R. To examine the impact of more modest, and biologically relevant, increases in RCAN1, we compared the mitochondrial network in induced pluripotent stem cells derived from individuals with Down syndrome to that of isogenic, disomic controls. Mitochondria were more fused, and O2 consumption was greater in the trisomic induced pluripotent stem cells; however, coupling efficiency and metabolic flexibility were compromised compared with disomic induced pluripotent stem cells. Depletion of RCAN1 from trisomic induced pluripotent stem cells was sufficient to normalize mitochondrial dynamics and function. CONCLUSIONS RCAN1 helps maintain a more interconnected mitochondrial network, and maintaining appropriate RCAN1 levels is important to human health and disease.
Collapse
Affiliation(s)
- Valentina Parra
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.).
| | - Francisco Altamirano
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Carolina P Hernández-Fuentes
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Dan Tong
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Victoriia Kyrychenko
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - David Rotter
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Zully Pedrozo
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Joseph A Hill
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Verónica Eisner
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Sergio Lavandero
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Jay W Schneider
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.)
| | - Beverly A Rothermel
- From the Advanced Center for Chronic Diseases and Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine (V.P., C.P.H.-F., Z.P., S.L.) and Institute of Biomedical Sciences, School of Medicine (Z.P.), University of Chile, Santiago; Department of Internal Medicine/Cardiology (V.P., F.A., D.T., V.K., D.R., Z.P., J.A.H., S.L., J.W.S., B.A.R.) and Department of Molecular Biology (V.K., J.A.H., B.A.R.), University of Texas Southwestern Medical Center, Dallas; and Department of Molecular and Cellular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago (V.E.).
| |
Collapse
|
36
|
Fructuoso M, Rachdi L, Philippe E, Denis RG, Magnan C, Le Stunff H, Janel N, Dierssen M. Increased levels of inflammatory plasma markers and obesity risk in a mouse model of Down syndrome. Free Radic Biol Med 2018; 114:122-130. [PMID: 28958596 DOI: 10.1016/j.freeradbiomed.2017.09.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/20/2017] [Accepted: 09/24/2017] [Indexed: 12/27/2022]
Abstract
Down syndrome (DS) is caused by the trisomy of human chromosome 21 and is the most common genetic cause of intellectual disability. In addition to the intellectual deficiencies and physical anomalies, DS individuals present a higher prevalence of obesity and subsequent metabolic disorders than healthy adults. There is increasing evidence from both clinical and experimental studies indicating the association of visceral obesity with a pro-inflammatory status and recent studies have reported that obese people with DS suffer from low-grade systemic inflammation. However, the link between adiposity and inflammation has not been explored in DS. Here we used Ts65Dn mice, a validated DS mouse model, for the study of obesity-related inflammatory markers. Ts65Dn mice presented increased energy intake, and a positive energy balance leading to increased adiposity (fat mass per body weight), but did not show overweight, which only was apparent upon high fat diet induced obesity. Trisomic mice also had fasting hyperglycemia and hypoinsulinemia, and normal incretin levels. Those trisomy-associated changes were accompanied by reduced ghrelin plasma levels and slightly but not significantly increased leptin levels. Upon a glucose load, Ts65Dn mice showed normal increase of incretins accompanied by over-responses of leptin and resistin, while maintaining the hyperglycemic and hypoinsulinemic phenotype. These changes in the adipoinsular axis were accompanied by increased plasma levels of inflammatory biomarkers previously correlated with obesity galectin-3 and HSP72, and reduced IL-6. Taken together, these results suggest that increased adiposity, and pro-inflammatory adipokines leading to low-grade inflammation are important players in the propensity to obesity in DS. We conclude that DS would be a case of impaired metabolic-inflammatory axis.
Collapse
Affiliation(s)
- M Fructuoso
- Cellular & Systems Neurobiology, Systems Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - L Rachdi
- INSERM U1016, Cochin Institute, Paris, France; CNRS UMR 8104, Paris, France; University of Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - E Philippe
- Université Paris Diderot, Sorbonne Paris Cité, Unité Biologie Fonctionnelle et Adaptative - CNRS UMR 8251, Paris, France
| | - R G Denis
- Université Paris Diderot, Sorbonne Paris Cité, Unité Biologie Fonctionnelle et Adaptative - CNRS UMR 8251, Paris, France
| | - C Magnan
- Université Paris Diderot, Sorbonne Paris Cité, Unité Biologie Fonctionnelle et Adaptative - CNRS UMR 8251, Paris, France
| | - H Le Stunff
- Université Paris Diderot, Sorbonne Paris Cité, Unité Biologie Fonctionnelle et Adaptative - CNRS UMR 8251, Paris, France; Université Paris Sud, France
| | - N Janel
- Université Paris Diderot, Sorbonne Paris Cité, Unité Biologie Fonctionnelle et Adaptative - CNRS UMR 8251, Paris, France
| | - M Dierssen
- Cellular & Systems Neurobiology, Systems Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain.
| |
Collapse
|
37
|
Liu C, Yu T, Xing Z, Jiang X, Li Y, Pao A, Mu J, Wallace PK, Stoica G, Bakin AV, Yu YE. Triplications of human chromosome 21 orthologous regions in mice result in expansion of megakaryocyte-erythroid progenitors and reduction of granulocyte-macrophage progenitors. Oncotarget 2017; 9:4773-4786. [PMID: 29435140 PMCID: PMC5797011 DOI: 10.18632/oncotarget.23463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/20/2017] [Indexed: 12/16/2022] Open
Abstract
Individuals with Down syndrome (DS) frequently have hematopoietic abnormalities, including transient myeloproliferative disorder and acute megakaryoblastic leukemia which are often accompanied by acquired GATA1 mutations that produce a truncated protein, GATA1s. The mouse has been used for modeling DS based on the syntenic conservation between human chromosome 21 (Hsa21) and three regions in the mouse genome located on mouse chromosome 10 (Mmu10), Mmu16 and Mmu17. To assess the impact of the dosage increase of Hsa21 gene orthologs on the hematopoietic system, we characterized the related phenotype in the Dp(10)1Yey/+;Dp(16)1Yey/+;Dp(17)1Yey/+ model which carries duplications spanning the entire Hsa21 orthologous regions on Mmu10, Mmu16 and Mmu17, and the Dp(10)1Yey/+;Dp(16)1Yey/+;Dp(17)1Yey/+;Gata1Yeym2 model which carries a Gata1s mutation we engineered. Both models exhibited anemia, macrocytosis, and myeloproliferative disorder. Similar to human DS, the megakaryocyte-erythrocyte progenitors (MEPs) and granulocyte-monocyte progenitors (GMPs) were significantly increased and reduced, respectively, in both models. The subsequent identification of all the aforementioned phenotypes in the Dp(16)1Yey/+ model suggests that the causative dosage sensitive gene(s) are in the Hsa21 orthologous region on Mmu16. Therefore, we reveal here for the first time that the human trisomy 21-associated major segmental chromosomal alterations in mice can lead to expanded MEP and reduced GMP populations, mimicking the dynamics of these myeloid progenitors in DS. These models will provide the critical systems for unraveling the molecular and cellular mechanism of DS-associated myeloproliferative disorder, and particularly for determining how human trisomy 21 leads to expansion of MEPs as well as how such an alteration leads to myeloproliferative disorder.
Collapse
Affiliation(s)
- Chunhong Liu
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Tao Yu
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.,Department of Medical Genetics, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhuo Xing
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Xiaoling Jiang
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Yichen Li
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Annie Pao
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Justin Mu
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Paul K Wallace
- Department of Flow and Image Cytometry, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - George Stoica
- Department of Pathobiology, Texas A&M University, College Station, TX 77843, USA
| | - Andrei V Bakin
- Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Y Eugene Yu
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.,Genetics, Genomics and Bioinformatics Program, State University of New York at Buffalo, Buffalo, NY 14263, USA
| |
Collapse
|
38
|
Peiris H, Keating DJ. The neuronal and endocrine roles of RCAN1 in health and disease. Clin Exp Pharmacol Physiol 2017; 45:377-383. [PMID: 29094385 DOI: 10.1111/1440-1681.12884] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/18/2017] [Accepted: 10/24/2017] [Indexed: 01/15/2023]
Abstract
The regulator of calcineurin 1 (RCAN1) was first discovered as a gene located on human chromosome 21, expressed in neurons and overexpressed in the brains of Down syndrome individuals. Increased expression of RCAN1 has been linked with not only Down syndrome-associated pathology but also an associated neurological disorder, Alzheimer's Disease, in which neuronal RCAN1 expression is also increased. RCAN1 has additionally been demonstrated to affect other cell types including endocrine cells, with links to the pathogenesis of β-cell dysfunction in type 2 diabetes. The primary functions of RCAN1 relate to the inhibition of the phosphatase calcineurin, and to the regulation of mitochondrial function. Various forms of cellular stress such as reactive oxygen species and hyperglycaemia cause transient increases in RCAN1 expression. The short term (hours to days) induction of RCAN1 expression is generally thought to have a protective effect by regulating the expression of pro-survival genes in multiple cell types, many of which are mediated via the calcineurin/NFAT transcriptional pathway. However, strong evidence also supports the notion that chronic (weeks-years) overexpression of RCAN1 has a detrimental effect on cells and that this may drive pathophysiological changes in neurons and endocrine cells linked to Down syndrome, Alzheimer's Disease and type 2 diabetes. Here we review the evidence related to these roles of RCAN1 in neurons and endocrine cells and their relationship to these human health disorders.
Collapse
Affiliation(s)
- Heshan Peiris
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, SA, Australia
| | - Damien J Keating
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
39
|
Minami T. Pathophysiology Derived from Down Syndrome Related Genes on the 21st Chromosome. YAKUGAKU ZASSHI 2017; 137:791-793. [PMID: 28674288 DOI: 10.1248/yakushi.16-00236-f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Takashi Minami
- Division of Vascular Biology, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo
| |
Collapse
|
40
|
Mathur A, Pandey VK, Kakkar P. PHLPP: a putative cellular target during insulin resistance and type 2 diabetes. J Endocrinol 2017; 233:R185-R198. [PMID: 28428363 DOI: 10.1530/joe-17-0081] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 04/20/2017] [Indexed: 12/29/2022]
Abstract
Progressive research in the past decade converges to the impact of PHLPP in regulating the cellular metabolism through PI3K/AKT inhibition. Aberrations in PKB/AKT signaling coordinates with impaired insulin secretion and insulin resistance, identified during T2D, obesity and cardiovascular disorders which brings in the relevance of PHLPPs in the metabolic paradigm. In this review, we discuss the impact of PHLPP isoforms in insulin signaling and its associated cellular events including mitochondrial dysfunction, DNA damage, autophagy and cell death. The article highlights the plausible molecular targets that share the role during insulin-resistant states, whose understanding can be extended into treatment responses to facilitate targeted drug discovery for T2D and allied metabolic syndromes.
Collapse
Affiliation(s)
- Alpana Mathur
- Herbal Research LaboratoryCSIR-Indian Institute of Toxicology Research, Lucknow, India
- Babu Banarasi Das UniversityBBD City, Lucknow, India
| | - Vivek Kumar Pandey
- Herbal Research LaboratoryCSIR-Indian Institute of Toxicology Research, Lucknow, India
- Academy of Scientific and Innovative ResearchCSIR-IITR, Lucknow, India
| | - Poonam Kakkar
- Herbal Research LaboratoryCSIR-Indian Institute of Toxicology Research, Lucknow, India
- Babu Banarasi Das UniversityBBD City, Lucknow, India
- Academy of Scientific and Innovative ResearchCSIR-IITR, Lucknow, India
| |
Collapse
|
41
|
Boles A, Kandimalla R, Reddy PH. Dynamics of diabetes and obesity: Epidemiological perspective. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1026-1036. [PMID: 28130199 PMCID: PMC5429876 DOI: 10.1016/j.bbadis.2017.01.016] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 01/17/2017] [Accepted: 01/23/2017] [Indexed: 02/08/2023]
Abstract
The purpose of this review article is to understand the current literature on obesity, diabetes and therapeutic avenues across the world. Diabetes is a chronic lifestyle condition that affects millions of people worldwide and it is a major health concern in our society. Diabetes and obesity are associated with various conditions, including non-modifiable and modifiable risk factors. Early detectable markers are not well established to detect pre-diabetes and as a result, it becomes diabetes. Several published epidemiological studies were assessed and the findings were summarized. Resources from published studies were used to identify criteria used for pre-diabetes, the role of diet in pre-diabetics and potential risks and characteristics associated with pre-diabetes. Preventive strategies are needed to combat diabetes. Individuals diagnosed with pre-diabetes need detailed education, need to fully understand the risk factors and have the ability to manage diabetes. Interventions exist that include chronic disease self-management programs, lifestyle interventions and pharmacological strategies. Obesity plays a large role in causing pre-diabetes and diabetes. Critical analysis of existing epidemiological research data suggests that additional research is needed to determine the efficacy of interventions. This article is part of a Special Issue entitled: Oxidative Stress and Mitochondrial Quality in Diabetes/Obesity and Critical Illness Spectrum of Diseases - edited by P. Hemachandra Reddy.
Collapse
Affiliation(s)
- Annette Boles
- Community Outreach and Education, 6630 S. Quaker Ave., Suite E, Lubbock, TX 79413, United States.
| | - Ramesh Kandimalla
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430-9424, United States; Department of Pharmacology & Neuroscience, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430-9424, United States.
| | - P Hemachandra Reddy
- Community Outreach and Education, 6630 S. Quaker Ave., Suite E, Lubbock, TX 79413, United States; Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430-9424, United States; Department of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430-9424, United States; Department of Pharmacology & Neuroscience, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430-9424, United States; Department of Neurology, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430-9424, United States; Speech, Language and Hearing Sciences Departments, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430-9424, United States; Department of Public Health, 3601 4th Street, MS 9424, Lubbock, TX 79430-9424, United States
| |
Collapse
|
42
|
Colvin KL, Yeager ME. What people with Down Syndrome can teach us about cardiopulmonary disease. Eur Respir Rev 2017; 26:26/143/160098. [DOI: 10.1183/16000617.0098-2016] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/13/2016] [Indexed: 12/19/2022] Open
Abstract
Down syndrome is the most common chromosomal abnormality among live-born infants. Through full or partial trisomy of chromosome 21, Down syndrome is associated with cognitive impairment, congenital malformations (particularly cardiovascular) and dysmorphic features. Immune disturbances in Down syndrome account for an enormous disease burden ranging from quality-of-life issues (autoimmune alopecia) to more serious health issues (autoimmune thyroiditis) and life-threatening issues (leukaemia, respiratory tract infections and pulmonary hypertension). Cardiovascular and pulmonary diseases account for ∼75% of the mortality seen in persons with Down syndrome. This review summarises the cardiovascular, respiratory and immune challenges faced by individuals with Down syndrome, and the genetic underpinnings of their pathobiology. We strongly advocate increased comparative studies of cardiopulmonary disease in persons with and without Down syndrome, as we believe these will lead to new strategies to prevent and treat diseases affecting millions of people worldwide.
Collapse
|
43
|
Duvoor C, Dendi VS, Marco A, Shekhawat NS, Chada A, Ravilla R, Musham CK, Mirza W, Chaudhury A. Commentary: ATP: The crucial component of secretory vesicles: Accelerated ATP/insulin exocytosis and prediabetes. Front Physiol 2017; 8:53. [PMID: 28210227 PMCID: PMC5288386 DOI: 10.3389/fphys.2017.00053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 01/19/2017] [Indexed: 02/03/2023] Open
Affiliation(s)
- Chitharanjan Duvoor
- Department of Endocrinology and Internal Medicine, University of Arkansas for Medical SciencesLittle Rock, AR, USA; GIM FoundationLittle Rock, AR, USA
| | - Vijaya S Dendi
- GIM FoundationLittle Rock, AR, USA; Department of Internal Medicine and Hospital Medicine, Christus Trinity Mother Frances HospitalTyler, TX, USA
| | - Asween Marco
- GIM FoundationLittle Rock, AR, USA; Department of Policy, University of Arkansas for Little RockLittle Rock, AR, USA
| | - Nawal S Shekhawat
- GIM FoundationLittle Rock, AR, USA; Tutwiler ClinicTutwiler, MS, USA
| | - Aditya Chada
- GIM FoundationLittle Rock, AR, USA; Department of Pulmonary and Critical Care Medicine, University of Arkansas for Medical SciencesLittle Rock, AR, USA
| | - Rahul Ravilla
- GIM FoundationLittle Rock, AR, USA; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical SciencesLittle Rock, AR, USA
| | - Chaitanya K Musham
- GIM FoundationLittle Rock, AR, USA; St. Vincent Infirmary (Catholic Health Initiative)Little Rock, AR, USA
| | | | | |
Collapse
|