1
|
Zuo X. Mitochondrial Imbalance in Down Syndrome: A Driver of Accelerated Brain Aging? Aging Dis 2025:AD.2025.0189. [PMID: 40249934 DOI: 10.14336/ad.2025.0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/06/2025] [Indexed: 04/20/2025] Open
Abstract
Down syndrome (DS), caused by trisomy of chromosome 21 (HSA21), is a complex condition associated with neurodevelopmental impairments and accelerated brain aging, often culminating in early-onset Alzheimer's disease (AD). Central to this accelerated aging is mitochondrial imbalance, characterized by disrupted energy metabolism, increased oxidative stress, impaired dynamics, and defective quality control mechanisms like mitophagy. These abnormalities exacerbate neuronal vulnerability, driving cognitive decline and neurodegeneration. This review examines the genetic and biochemical underpinnings of mitochondrial dysfunction in DS, with a focus on the role of HSA21-encoded genes. We also highlight how mitochondrial dysfunction, amplified by oxidative stress and HSA21 gene dosage effects, converges with cellular senescence and neuroinflammation to accelerate Alzheimer-like pathology and brain aging in DS. Finally, we discuss emerging therapeutic strategies targeting mitochondrial pathways, which hold promise for mitigating neurodegenerative phenotypes and improving outcomes in DS.
Collapse
|
2
|
Valdor R, Martinez-Vicente M. The Role of Chaperone-Mediated Autophagy in Tissue Homeostasis and Disease Pathogenesis. Biomedicines 2024; 12:257. [PMID: 38397859 PMCID: PMC10887052 DOI: 10.3390/biomedicines12020257] [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: 12/07/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
Chaperone-mediated autophagy (CMA) is a selective proteolytic pathway in the lysosomes. Proteins are recognized one by one through the detection of a KFERQ motif or, at least, a KFERQ-like motif, by a heat shock cognate protein 70 (Hsc70), a molecular chaperone. CMA substrates are recognized and delivered to a lysosomal CMA receptor, lysosome-associated membrane protein 2A (LAMP-2A), the only limiting component of this pathway, and transported to the lysosomal lumen with the help of another resident chaperone HSp90. Since approximately 75% of proteins are reported to have canonical, phosphorylation-generated, or acetylation-generated KFERQ motifs, CMA maintains intracellular protein homeostasis and regulates specific functions in the cells in different tissues. CMA also regulates physiologic functions in different organs, and is then implicated in disease pathogenesis related to aging, cancer, and the central nervous and immune systems. In this minireview, we have summarized the most important findings on the role of CMA in tissue homeostasis and disease pathogenesis, updating the recent advances for this Special Issue.
Collapse
Affiliation(s)
- Rut Valdor
- Immunology-Cell Therapy and Hematopoietic Transplant Group, Department of Biochemistry and Molecular Biology B, University of Murcia (UMU), 30100 Murcia, Spain
- Unit of Autophagy, Immune Response and Tolerance in Pathologic Processes, Biomedical Research Institute of Murcia-Pascual Parrilla (IMIB), 30120 Murcia, Spain
| | - Marta Martinez-Vicente
- Autophagy and Lysosomal Dysfunction Lab, Neurodegenerative Diseases Research Group, Vall d’Hebron Research Institute—CIBERNED, 08035 Barcelona, Spain
| |
Collapse
|
3
|
Kotchetkov P, Blakeley N, Lacoste B. Involvement of brain metabolism in neurodevelopmental disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 173:67-113. [PMID: 37993180 DOI: 10.1016/bs.irn.2023.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Neurodevelopmental disorders (NDDs) affect a significant portion of the global population and have a substantial social and economic impact worldwide. Most NDDs manifest in early childhood and are characterized by deficits in cognition, communication, social interaction and motor control. Due to a limited understanding of the etiology of NDDs, current treatment options primarily focus on symptom management rather than on curative solutions. Moreover, research on NDDs is problematic due to its reliance on a neurocentric approach. However, recent studies are broadening the scope of research on NDDs, to include dysregulations within a diverse network of brain cell types, including vascular and glial cells. This review aims to summarize studies from the past few decades on potential new contributions to the etiology of NDDs, with a special focus on metabolic signatures of various brain cells. In particular, we aim to convey how the metabolic functions are intimately linked to the onset and/or progression of common NDDs such as autism spectrum disorders, fragile X syndrome, Rett syndrome and Down syndrome.
Collapse
Affiliation(s)
- Pavel Kotchetkov
- Neuroscience Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Nicole Blakeley
- Neuroscience Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, ON, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
4
|
Elangovan A, Babu HWS, Iyer M, Gopalakrishnan AV, Vellingiri B. Untangle the mystery behind DS-associated AD - Is APP the main protagonist? Ageing Res Rev 2023; 87:101930. [PMID: 37031726 DOI: 10.1016/j.arr.2023.101930] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023]
Abstract
Amyloid precursor protein profusion in Trisomy 21, also called Down Syndrome (DS), is rooted in the genetic determination of Alzheimer's disease (AD). With the recent development in patient care, the life expectancy of DS patients has gradually increased, leading to the high prospect of AD development, consequently leading to the development of plaques of amyloid proteins and neurofibrillary tangles made of tau by the fourth decade of the patient leading to dementia. The altered gene expression resulted in cellular dysfunction due to impairment of autophagy, mitochondrial and lysosomal dysfunction, and copy number variation controlled by the additional genes in Trisomy 21. The cognitive impairment and mechanistic insights underlying DS-AD conditions have been reviewed in this article. Some recent findings regarding biomarkers and therapeutics of DS-AD conditions were highlighted in this review.
Collapse
Affiliation(s)
- Ajay Elangovan
- Stem cell and Regenerative Medicine/ Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, Punjab, India; Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - Harysh Winster Suresh Babu
- Stem cell and Regenerative Medicine/ Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, Punjab, India; Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - Mahalaxmi Iyer
- Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to be University), Coimbatore-641021, India
| | | | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/ Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, Punjab, India; Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India.
| |
Collapse
|
5
|
Tan KL, Lee HC, Cheah PS, Ling KH. Mitochondrial Dysfunction in Down Syndrome: From Pathology to Therapy. Neuroscience 2023; 511:1-12. [PMID: 36496187 DOI: 10.1016/j.neuroscience.2022.12.003] [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: 06/07/2022] [Revised: 10/07/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022]
Abstract
Mitochondrial dysfunctions have been described in Down syndrome (DS) caused by either partial or full trisomy of chromosome 21 (HSA21). Mitochondria play a crucial role in various vital functions in eukaryotic cells, especially in energy production, calcium homeostasis and programmed cell death. The function of mitochondria is primarily regulated by genes encoded in the mitochondrion and nucleus. Many genes on HSA21 are involved in oxidative phosphorylation (OXPHOS) and regulation of mitochondrial functions. This review highlights the HSA21 dosage-sensitive nuclear-encoded mitochondrial genes associated with overexpression-related phenotypes seen in DS. This includes impaired mitochondrial dynamics, structural defects and dysregulated bioenergetic profiles such as OXPHOS deficiency and reduced ATP production. Various therapeutic approaches for modulating energy deficits in DS, effects and molecular mechanism of gene therapy and drugs that exert protective effects through modulation of mitochondrial function and attenuation of oxidative stress in DS cells were discussed. It is prudent that improving DS pathophysiological conditions or quality of life may be feasible by targeting something as simple as cellular mitochondrial biogenesis and function.
Collapse
Affiliation(s)
- Kai-Leng Tan
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia; Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Han-Chung Lee
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia; Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Pike-See Cheah
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia; Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - King-Hwa Ling
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia; Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia; Department of Genetics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Choi C, Kim H, Oh J, Park C, Kim M, Kim CS, Park J. DSCR1 deficiency ameliorates the Aβ pathology of Alzheimer's disease by enhancing microglial activity. Life Sci Alliance 2023; 6:6/2/e202201556. [PMID: 36450444 PMCID: PMC9713304 DOI: 10.26508/lsa.202201556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 12/02/2022] Open
Abstract
Microglial phagocytosis and clearance are important for the removal of amyloid-β (Aβ) plaques in Alzheimer's disease (AD). Chronic exposure of microglia to Aβ plaques leads to microglial metabolic dysfunction, and dysregulation of microglia can accelerate the deposition of Aβ plaques and cause learning and memory impairment. Thus, regulating microglial Aβ clearance is crucial for the development of therapeutics for AD-related dementia. Here, Down syndrome critical region 1 (DSCR1) deficiency ameliorated Aβ plaque deposition in the 5xFAD mouse model of AD by altering microglial activity; however, the Aβ synthesis pathway was not affected. DSCR1 deficiency improved spatial learning and memory impairment in 5xFAD mice. Furthermore, DSCR1-deficient microglia exhibited accelerated lysosomal degradation of Aβ after phagocytosis, and BV2 cells with stable knockdown of DSCR1 demonstrated enhanced lysosomal activity. RNA-sequencing analysis showed that the transcriptional signatures associated with responses to IFN-γ were significantly up-regulated in DSCR1-knockdown BV2 cells treated with Aβ. Our data strongly suggest that DSCR1 is a critical mediator of microglial degradation of amyloid plaques and a new potential microglial therapeutic target in AD.
Collapse
Affiliation(s)
- Chiyeol Choi
- Department of Biological Sciences, College of Information and Bioengineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Hyerin Kim
- Department of Biological Sciences, College of Information and Bioengineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jiyoung Oh
- Department of Biological Sciences, College of Information and Bioengineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Chanho Park
- Department of Biological Sciences, College of Information and Bioengineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Min Kim
- Department of Biological Sciences, College of Information and Bioengineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Chu-Sook Kim
- Department of Biological Sciences, College of Information and Bioengineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jiyoung Park
- Department of Biological Sciences, College of Information and Bioengineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| |
Collapse
|
7
|
Li Y, Costiniti V, Souza Bomfim GH, Neginskaya M, Son GY, Rothermel B, Pavlov E, Lacruz RS. Overexpression of RCAN1, a Gene on Human Chromosome 21, Alters Cell Redox and Mitochondrial Function in Enamel Cells. Cells 2022; 11:3576. [PMID: 36429004 PMCID: PMC9688881 DOI: 10.3390/cells11223576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
The regulator of calcineurin (RCAN1) has been implicated in the pathogenesis of Down syndrome (DS). Individuals with DS show dental abnormalities for unknown reasons, and RCAN1 levels have been found to be elevated in several tissues of DS patients. A previous microarray analysis comparing cells of the two main formative stages of dental enamel, secretory and maturation, showed a significant increase in RCAN1 expression in the latter. Because the function of RCAN1 during enamel formation is unknown, there is no mechanistic evidence linking RCAN1 with the dental anomalies in individuals with DS. We investigated the role of RCAN1 in enamel by overexpressing RCAN1 in the ameloblast cell line LS8 (LS8+RCAN1). We first confirmed that RCAN1 is highly expressed in maturation stage ameloblasts by qRT-PCR and used immunofluorescence to show its localization in enamel-forming ameloblasts. We then analyzed cell redox and mitochondrial bioenergetics in LS8+RCAN1 cells because RCAN1 is known to impact these processes. We show that LS8+RCAN1 cells have increased reactive oxygen species (ROS) and decreased mitochondrial bioenergetics without changes in the expression of the complexes of the electron transport chain, or in NADH levels. However, LS8+RCAN1 cells showed elevated mitochondrial Ca2+ uptake and decreased expression of several enamel genes essential for enamel formation. These results provide insight into the role of RCAN1 in enamel and suggest that increased RCAN1 levels in the ameloblasts of individuals with DS may impact enamel formation by altering both the redox environment and mitochondrial function, as well as decreasing the expression of enamel-specific genes.
Collapse
Affiliation(s)
- Yi Li
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Veronica Costiniti
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Guilherme H. Souza Bomfim
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Maria Neginskaya
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Ga-Yeon Son
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Beverly Rothermel
- Department of Internal Medicine and Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Evgeny Pavlov
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Rodrigo S. Lacruz
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| |
Collapse
|
8
|
Lao M, Zhang X, Yang H, Bai X, Liang T. RCAN1-mediated calcineurin inhibition as a target for cancer therapy. Mol Med 2022; 28:69. [PMID: 35717152 PMCID: PMC9206313 DOI: 10.1186/s10020-022-00492-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/26/2022] [Indexed: 11/10/2022] Open
Abstract
Cancer is the leading cause of mortality worldwide. Regulator of calcineurin 1 (RCAN1), as a patent endogenous inhibitor of calcineurin, plays crucial roles in the pathogenesis of cancers. Except for hypopharyngeal and laryngopharynx cancer, high expression of RCAN1 inhibits tumor progression. Molecular antitumor functions of RCAN1 are largely dependent on calcineurin. In this review, we highlight current research on RCAN1 characteristics, and the interaction between RCAN1 and calcineurin. Moreover, the dysregulation of RCAN1 in various cancers is reviewed, and the potential of targeting RCAN1 as a new therapeutic approach is discussed.
Collapse
Affiliation(s)
- Mengyi Lao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China
| | - Xiaozhen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China
| | - Hanshen Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China.,Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China. .,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China. .,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China. .,Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China.
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310009, Zhejiang, China. .,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China. .,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China. .,Zhejiang Provincial Clinical Research Center for the Study of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310009, Zhejiang, China. .,Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
9
|
Wong H, Buck JM, Borski C, Pafford JT, Keller BN, Milstead RA, Hanson JL, Stitzel JA, Hoeffer CA. RCAN1 knockout and overexpression recapitulate an ensemble of rest-activity and circadian disruptions characteristic of Down syndrome, Alzheimer's disease, and normative aging. J Neurodev Disord 2022; 14:33. [PMID: 35610565 PMCID: PMC9128232 DOI: 10.1186/s11689-022-09444-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 05/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Regulator of calcineurin 1 (RCAN1) is overexpressed in Down syndrome (DS), but RCAN1 levels are also increased in Alzheimer's disease (AD) and normal aging. AD is highly comorbid among individuals with DS and is characterized in part by progressive neurodegeneration that resembles accelerated aging. Importantly, abnormal RCAN1 levels have been demonstrated to promote memory deficits and pathophysiology that appear symptomatic of DS, AD, and aging. Anomalous diurnal rest-activity patterns and circadian rhythm disruptions are also common in DS, AD, and aging and have been implicated in facilitating age-related cognitive decline and AD progression. However, no prior studies have assessed whether RCAN1 dysregulation may also promote the age-associated alteration of rest-activity profiles and circadian rhythms, which could in turn contribute to neurodegeneration in DS, AD, and aging. METHODS The present study examined the impacts of RCAN1 deficiency and overexpression on the photic entrainment, circadian periodicity, intensity and distribution, diurnal patterning, and circadian rhythmicity of wheel running in young (3-6 months old) and aged (9-14 months old) mice of both sexes. RESULTS We found that daily RCAN1 levels in the hippocampus and suprachiasmatic nucleus (SCN) of light-entrained young mice are generally constant and that balanced RCAN1 expression is necessary for normal circadian locomotor activity rhythms. While the light-entrained diurnal period was unaltered, RCAN1-null and RCAN1-overexpressing mice displayed lengthened endogenous (free-running) circadian periods like mouse models of AD and aging. In light-entrained young mice, RCAN1 deficiency and overexpression also recapitulated the general hypoactivity, diurnal rest-wake pattern fragmentation, and attenuated amplitudes of circadian activity rhythms reported in DS, preclinical and clinical AD, healthily aging individuals, and rodent models thereof. Under constant darkness, RCAN1-null and RCAN1-overexpressing mice displayed altered locomotor behavior indicating circadian clock dysfunction. Using the Dp(16)1Yey/+ (Dp16) mouse model for DS, which expresses three copies of Rcan1, we found reduced wheel running activity and rhythmicity in both light-entrained and free-running young Dp16 mice like young RCAN1-overexpressing mice. Critically, these diurnal and circadian deficits were rescued in part or entirely by restoring Rcan1 to two copies in Dp16 mice. We also found that RCAN1 deficiency but not RCAN1 overexpression altered protein levels of the clock gene Bmal1 in the SCN. CONCLUSIONS Collectively, this study's findings suggest that both loss and aberrant gain of RCAN1 precipitate anomalous light-entrained diurnal and circadian activity patterns emblematic of DS, AD, and possibly aging.
Collapse
Affiliation(s)
- Helen Wong
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Jordan M Buck
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Curtis Borski
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Jessica T Pafford
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Bailey N Keller
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
| | - Ryan A Milstead
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Jessica L Hanson
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA
| | - Charles A Hoeffer
- Institute for Behavioral Genetics, University of Colorado Boulder, 1480 30th Street, Boulder, CO, 80309-0447, USA.
- Department of Integrative Physiology, University of Colorado, Boulder, CO, 80303, USA.
- Linda Crnic Institute, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
10
|
Noll C, Kandiah J, Moroy G, Gu Y, Dairou J, Janel N. Catechins as a Potential Dietary Supplementation in Prevention of Comorbidities Linked with Down Syndrome. Nutrients 2022; 14:2039. [PMID: 35631180 PMCID: PMC9147372 DOI: 10.3390/nu14102039] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 11/24/2022] Open
Abstract
Plant-derived polyphenols flavonoids are increasingly being recognized for their medicinal potential. These bioactive compounds derived from plants are gaining more interest in ameliorating adverse health risks because of their low toxicity and few side effects. Among them, therapeutic approaches demonstrated the efficacy of catechins, a major group of flavonoids, in reverting several aspects of Down syndrome, the most common genomic disorder that causes intellectual disability. Down syndrome is characterized by increased incidence of developing Alzheimer's disease, obesity, and subsequent metabolic disorders. In this focused review, we examine the main effects of catechins on comorbidities linked with Down syndrome. We also provide evidence of catechin effects on DYRK1A, a dosage-sensitive gene encoding a protein kinase involved in brain defects and metabolic disease associated with Down syndrome.
Collapse
Affiliation(s)
- Christophe Noll
- Division of Endocrinology, Department of Medicine, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada;
| | - Janany Kandiah
- Unité de Biologie Fonctionnelle et Adaptative, UMR 8251 CNRS, Université Paris Cité, F-75013 Paris, France; (J.K.); (Y.G.)
| | - Gautier Moroy
- Unité de Biologie Fonctionnelle et Adaptative, INSERM CNRS, Université Paris Cité, F-75013 Paris, France;
| | - Yuchen Gu
- Unité de Biologie Fonctionnelle et Adaptative, UMR 8251 CNRS, Université Paris Cité, F-75013 Paris, France; (J.K.); (Y.G.)
| | - Julien Dairou
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, UMR 8601 CNRS, Université Paris Cité, F-75006 Paris, France;
| | - Nathalie Janel
- Unité de Biologie Fonctionnelle et Adaptative, UMR 8251 CNRS, Université Paris Cité, F-75013 Paris, France; (J.K.); (Y.G.)
| |
Collapse
|
11
|
McKetney J, Jenkins CC, Minogue C, Mach PM, Hussey EK, Glaros TG, Coon J, Dhummakupt ES. Proteomic and metabolomic profiling of acute and chronic stress events associated with military exercises. Mol Omics 2021; 18:279-295. [PMID: 34860218 DOI: 10.1039/d1mo00271f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
By characterizing physiological changes that occur in warfighters during simulated combat, we can start to unravel the key biomolecular components that are linked to physical and cognitive performance. Viable field-based sensors for the warfighter must be rapid and noninvasive. In an effort to facilitate this, we applied a multiomics pipeline to characterize the stress response in the saliva of warfighters to correlate biomolecular changes with overall performance and health. In this study, two different stress models were observed - one of chronic stress and one of acute stress. In both models, significant perturbations in the immune, metabolic, and protein manufacturing/processing systems were observed. However, when differentiating between stress models, specific metabolites associated with the "fight or flight" response and protein folding were seen to be discriminate of the acute stress model.
Collapse
Affiliation(s)
- Justin McKetney
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, 53706, USA. .,National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA
| | - Conor C Jenkins
- DEVCOM Chemical Biological Center, Aberdeen Proving Grounds, MD 21010, USA.
| | - Catie Minogue
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, 53706, USA. .,National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA
| | - Phillip M Mach
- DEVCOM Chemical Biological Center, Aberdeen Proving Grounds, MD 21010, USA.
| | - Erika K Hussey
- DEVCOM Soldier Center, Natick, MA 01760, USA.,Defense Innovation Unit, Mountain View, CA 94043, USA
| | - Trevor G Glaros
- DEVCOM Chemical Biological Center, Aberdeen Proving Grounds, MD 21010, USA. .,Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Joshua Coon
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, 53706, USA. .,National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA.,Morgridge Institute for Research, Madison, WI 53515, USA.,Department of Chemistry, University of Wisconsin, Madison, WI 53706, USA
| | | |
Collapse
|
12
|
Zheng H, Zhu H, Liu X, Huang X, Huang A, Huang Y. Mitophagy in Diabetic Cardiomyopathy: Roles and Mechanisms. Front Cell Dev Biol 2021; 9:750382. [PMID: 34646830 PMCID: PMC8503602 DOI: 10.3389/fcell.2021.750382] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/06/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular disease is the leading complication of diabetes mellitus (DM), and diabetic cardiomyopathy (DCM) is a major cause of mortality in diabetic patients. Multiple pathophysiologic mechanisms, including myocardial insulin resistance, oxidative stress and inflammation, are involved in the development of DCM. Recent studies have shown that mitochondrial dysfunction makes a substantial contribution to the development of DCM. Mitophagy is a type of autophagy that takes place in dysfunctional mitochondria, and it plays a key role in mitochondrial quality control. Although the precise molecular mechanisms of mitophagy in DCM have yet to be fully clarified, recent findings imply that mitophagy improves cardiac function in the diabetic heart. However, excessive mitophagy may exacerbate myocardial damage in patients with DCM. In this review, we aim to provide a comprehensive overview of mitochondrial quality control and the dual roles of mitophagy in DCM. We also propose that a balance between mitochondrial biogenesis and mitophagy is essential for the maintenance of cellular metabolism in the diabetic heart.
Collapse
Affiliation(s)
- Haoxiao Zheng
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
| | - Hailan Zhu
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
| | - Xinyue Liu
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
| | - Xiaohui Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
| | - Anqing Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
| | - Yuli Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation Research, Guangzhou, China
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
13
|
Yang Y, Zhou X, Liu X, Song R, Gao Y, Wang S. Implications of FBXW7 in Neurodevelopment and Neurodegeneration: Molecular Mechanisms and Therapeutic Potential. Front Cell Neurosci 2021; 15:736008. [PMID: 34512273 PMCID: PMC8424092 DOI: 10.3389/fncel.2021.736008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/04/2021] [Indexed: 11/25/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) mediated protein degradation is crucial to maintain quantitive and functional homeostasis of diverse proteins. Balanced cellular protein homeostasis controlled by UPS is fundamental to normal neurological functions while impairment of UPS can also lead to some neurodevelopmental and neurodegenerative disorders. Functioning as the substrate recognition component of the SCF-type E3 ubiquitin ligase, FBXW7 is essential to multiple aspects of cellular processes via targeting a wide range of substrates for proteasome-mediated degradation. Accumulated evidence shows that FBXW7 is fundamental to neurological functions and especially implicated in neurodevelopment and the nosogenesis of neurodegeneration. In this review, we describe general features of FBXW7 gene and proteins, and mainly present recent findings that highlight the vital roles and molecular mechanisms of FBXW7 in neurodevelopment such as neurogenesis, myelination and cerebral vasculogenesis and in the pathogenesis of some typical neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease and Huntington’s disease. Additionally, we also provide a prospect on focusing FBXW7 as a potential therapeutic target to rescue neurodevelopmental and neurodegenerative impairment.
Collapse
Affiliation(s)
- Yu Yang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Xuan Zhou
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China.,Research Center for Quality of Life and Applied Psychology, School of Humanities and Management, Guangdong Medical University, Dongguan, China
| | - Xinpeng Liu
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Ruying Song
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Yiming Gao
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Shuai Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| |
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: 3.8] [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
|
Epigallocatechin-3-Gallate Plus Omega-3 Restores the Mitochondrial Complex I and F 0F 1-ATP Synthase Activities in PBMCs of Young Children with Down Syndrome: A Pilot Study of Safety and Efficacy. Antioxidants (Basel) 2021; 10:antiox10030469. [PMID: 33809669 PMCID: PMC8002266 DOI: 10.3390/antiox10030469] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/05/2021] [Accepted: 03/14/2021] [Indexed: 12/15/2022] Open
Abstract
Down syndrome (DS) is a major genetic cause of intellectual disability. DS pathogenesis has not been fully elucidated, and no specific pharmacological therapy is available. DYRK1A overexpression, oxidative stress and mitochondrial dysfunction were described in trisomy 21. Epigallocatechin-3-gallate (EGCG) is a multimodal nutraceutical with antioxidant properties. EGCG inhibits DYRK1A overexpression and corrects DS mitochondrial dysfunction in vitro. The present study explores safety profiles in DS children aged 1–8 years treated with EGCG (10 mg/kg/die, suspended in omega-3, per os, in fasting conditions, for 6 months) and EGCG efficacy in restoring mitochondrial complex I and F0F1-ATP synthase (complex V) deficiency, assessed on PBMCs. The Griffiths Mental Developmental Scales—Extended Revised (GMDS-ER) was used for developmental profiling. Results show that decaffeinated EGCG (>90%) plus omega-3 is safe in DS children and effective in reverting the deficit of mitochondrial complex I and V activities. Decline of plasma folates was observed in 21% of EGCG-treated patients and should be carefully monitored. GMDS-ER scores did not show differences between the treated group compared to the DS control group. In conclusion, EGCG plus omega-3 can be safely administered under medical supervision in DS children aged 1–8 years to normalize mitochondria respiratory chain complex activities, while results on the improvement of developmental performance are still inconclusive.
Collapse
|
16
|
Lin HQ, Dai SH, Liu WC, Lin X, Yu BT, Chen SB, Liu S, Ling H, Tang J. Effects of prolonged cold-ischemia on autophagy in the graft lung in a rat orthotopic lung transplantation model. Life Sci 2021; 268:118820. [PMID: 33278393 DOI: 10.1016/j.lfs.2020.118820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Ischemia-reperfusion (I/R) injury causes present challenges in the field of graft transplantation which is also a major contributor to early graft dysfunction or failure after organ transplantation. The study focuses on the effects of prolonged cold-ischemia (CI) on the autophagic activity in the graft lung in a rat orthotopic lung transplantation model. MATERIAL AND METHODS Donor lungs were preserved under CI conditions for different periods. An orthotopic lung transplantation model was developed, and the lung tissues from donor lungs subjected to CI preservation and reperfusion were harvested. We evaluated the effects of different CI periods on autophagy, reactive oxygen species (ROS) and glucose consumption. Additionally, the mechanism by which prolonged CI affected autophagy was investigated through determination of the molecules related to the mTOR pathway after treatment with 3-Methyladenine (3-MA), rapamycin and an adenosine triphosphate (ATP) synthase inhibitor oligomycin (OM). RESULTS Prolonged CI led to increased activities of key glycolytic enzymes, glucose consumption and lactic acid production. Autophagy, ROS and glucose consumption were induced in the graft lung after I/R, which reached peak levels after 6 h and was gradually decreased. Most importantly, the perfusion treatment of 3-MA or OM decreased ROS level and autophagy, but increased the extent of mTOR phosphorylation, while the perfusion treatment of rapamycin induced ROS and autophagy. CONCLUSION Taken together, autophagy mediated by a prolonged CI preservation affects the glucose consumption and ROS production in the graft lung via the mTOR signaling pathway.
Collapse
Affiliation(s)
- Hui-Qing Lin
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, PR China
| | - Shao-Hua Dai
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Wei-Cheng Liu
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Xiang Lin
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Ben-Tong Yu
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Shi-Biao Chen
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Sheng Liu
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Hua Ling
- Department of Nursing, the First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China.
| | - Jian Tang
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China.
| |
Collapse
|
17
|
A transient decrease in mitochondrial activity contributes to establish the ganglion cell fate in retina adapted for high acuity vision. Dev Biol 2020; 469:96-110. [PMID: 33141037 DOI: 10.1016/j.ydbio.2020.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/17/2022]
Abstract
Although the plan of the retina is well conserved in vertebrates, there are considerable variations in cell type diversity and number, as well as in the organization and properties of the tissue. The high ratios of retinal ganglion cells (RGCs) to cones in primate fovea and bird retinas favor neural circuits essential for high visual acuity and color vision. The role that cell metabolism could play in cell fate decision during embryonic development of the nervous system is still largely unknown. Here, we describe how subtle changes of mitochondrial activity along the pathway converting uncommitted progenitors into newborn RGCs increase the recruitment of RGC-fated progenitors. ATOH7, a proneural protein dedicated to the production of RGCs in vertebrates, activates transcription of the Hes5.3 gene in pre-committed progenitors. The HES5.3 protein, in turn, regulates a transient decrease in mitochondrial activity via the retinoic acid signaling pathway few hours before cell commitment. This metabolic shift lengthens the progression of the ultimate cell cycle and is a necessary step for upregulating Atoh7 and promoting RGC differentiation.
Collapse
|
18
|
Martínez-Cué C, Rueda N. Signalling Pathways Implicated in Alzheimer's Disease Neurodegeneration in Individuals with and without Down Syndrome. Int J Mol Sci 2020; 21:E6906. [PMID: 32962300 PMCID: PMC7555886 DOI: 10.3390/ijms21186906] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Down syndrome (DS), the most common cause of intellectual disability of genetic origin, is characterized by alterations in central nervous system morphology and function that appear from early prenatal stages. However, by the fourth decade of life, all individuals with DS develop neuropathology identical to that found in sporadic Alzheimer's disease (AD), including the development of amyloid plaques and neurofibrillary tangles due to hyperphosphorylation of tau protein, loss of neurons and synapses, reduced neurogenesis, enhanced oxidative stress, and mitochondrial dysfunction and neuroinflammation. It has been proposed that DS could be a useful model for studying the etiopathology of AD and to search for therapeutic targets. There is increasing evidence that the neuropathological events associated with AD are interrelated and that many of them not only are implicated in the onset of this pathology but are also a consequence of other alterations. Thus, a feedback mechanism exists between them. In this review, we summarize the signalling pathways implicated in each of the main neuropathological aspects of AD in individuals with and without DS as well as the interrelation of these pathways.
Collapse
Affiliation(s)
- Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, 39011 Santander, Spain;
| | | |
Collapse
|
19
|
Molecular mechanisms of interplay between autophagy and metabolism in cancer. Life Sci 2020; 259:118184. [PMID: 32763290 DOI: 10.1016/j.lfs.2020.118184] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022]
Abstract
Autophagy is an essential mechanism of cellular degradation, a way to protect the cells under stress conditions, such as deprivation of nutrients, growth factors and cellular damage. However, in normal physiology autophagy plays a significant role in cancer cells. Current research is in progress to understand how autophagy and cancer cells go hand in hand to support cancer cell progression. The important aspect in cancer and autophagy is the interdependence of autophagy in the survival and progression of cancer cells. Autophagy is known to be a major cause of chemotherapeutic resistance in various cancer cell types. Therefore, inhibition of autophagy as an effective therapeutic approach is being actively studied and tested in clinical studies. Multiple metabolic pathways are linked with autophagy that could potentially be a significant target for chemotherapeutic strategy. The comprehension of the interconnection of autophagy with cancer metabolism can pave a novel findings for effective combinatorial therapeutic strategies.
Collapse
|
20
|
Xue Z, Zhang Y, Liu Y, Zhang C, Shen XD, Gao F, Busuttil RW, Zheng S, Kupiec-Weglinski JW, Ji H. PACAP neuropeptide promotes Hepatocellular Protection via CREB-KLF4 dependent autophagy in mouse liver Ischemia Reperfusion Injury. Am J Cancer Res 2020; 10:4453-4465. [PMID: 32292507 PMCID: PMC7150481 DOI: 10.7150/thno.42354] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
Organ ischemia reperfusion injury (IRI), associated with acute hepatocyte death, remains an unresolved problem in clinical orthotopic liver transplantation (OLT). Autophagy, an intracellular self-digesting progress, is responsible for cell reprograming required to regain post-stress homeostasis. Methods: Here, we analyzed the cytoprotective mechanism of pituitary adenylate cyclase-activating polypeptide (PACAP)-promoted hepatocellular autophagy in a clinically relevant mouse model of extended hepatic cold storage (4 °C UW solution for 20 h) followed by syngeneic OLT. Results: In contrast to 41.7% of liver graft failure by day 7 post-transplant in control group, PACAP treatment significantly improved graft survival (91.7% by day 14), and promoted autophagy-associated regeneration programs in OLT. In parallel in vitro studies, PACAP-enhanced autophagy ameliorated cellular damage (LDH/ALT levels), and diminished necrosis in H2O2-stressed primary hepatocytes. Interestingly, PACAP not only induced nuclear cAMP response element-binding protein (CREB), but also triggered reprogramming factor Kruppel-like factor 4 (KLF4) expression in IR-stressed OLT. Indeed, CREB inhibition attenuated hepatic autophagy and recreated hepatocellular injury in otherwise PACAP-protected livers. Furthermore, CREB inhibition suppressed PACAP-induced KLF4 expression, whereas KLF4 blockade abolished PACAP-promoted autophagy and neutralized PACAP-mediated hepatoprotection both in vivo and in vitro. Conclusion: Current study documents the essential neural regulation of PACAP-promoted autophagy in hepatocellular homeostasis in OLT, which provides the emerging therapeutic principle to combat hepatic IRI in OLT.
Collapse
|
21
|
Wang H, Zhang T, Ge X, Chen J, Zhao Y, Fu J. Parkin overexpression attenuates Aβ-induced mitochondrial dysfunction in HEK293 cells by restoring impaired mitophagy. Life Sci 2020; 244:117322. [PMID: 31958419 DOI: 10.1016/j.lfs.2020.117322] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 10/25/2022]
Abstract
AIMS Mitochondrial dysfunction is an early prominent feature of Alzheimer's disease (AD). In the present study, we sought to investigate whether defective mitophagy is tightly related to amyloid-β (Aβ)-induced mitochondrial dysfunction. MAIN METHODS Immunofluorescence, western blot and transmission electron microscopy were used to examine mitophagy. Mitochondrial membrane potential was assessed using the JC-1 dye. Mitochondrial ROS was detected using MitoSOX™ Red staining. KEY FINDINGS Aβ induced mitochondrial dysfunction in HEK293 cells. Moreover, Aβ induced an increase in parkin translocation to mitochondria and led to a drastic reduction in cytosolic parkin. Furthermore, Aβ-treated cells displayed a microtubule-associated protein 1 light chain 3 (LC3) punctate pattern and elevated mitochondrial LC3-II levels, suggesting the upregulation of mitophagy. Notably, Aβ induced the accumulation of mitochondrial p62, which was associated with impaired mitophagy. In addition, Aβ-treated cells exhibited fragmented or swollen mitochondria with severely decreased cristae. We then investigated whether overexpression of parkin could protect cells against Aβ-induced mitochondrial dysfunction. Interestingly, parkin overexpression inhibited Aβ-induced mitochondrial dysfunction. Besides, parkin overexpression increased cytosolic and mitochondrial parkin levels as well as mitochondrial LC3-II levels in Aβ-treated cells. Additionally, parkin overexpression reversed the accumulation of p62 in mitochondria, indicating that parkin overexpression restored impaired mitophagy in Aβ-treated cells. Importantly, parkin overexpression remarkably reversed Aβ-induced mitochondrial fragmentation. SIGNIFICANCE Our data demonstrate that overexpression of parkin ameliorates impaired mitophagy and promotes the removal of damaged mitochondria in Aβ-treated cells, indicating that upregulation of parkin-mediated mitophagy may be a potential strategy for the therapy of AD.
Collapse
Affiliation(s)
- Hongmei Wang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ting Zhang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xuhua Ge
- Department of General Medicine, Yangpu Hospital Affiliated to Tongji University, Shanghai, China
| | - Jingjiong Chen
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yuwu Zhao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Jianliang Fu
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
22
|
Gomez W, Morales R, Maracaja-Coutinho V, Parra V, Nassif M. Down syndrome and Alzheimer's disease: common molecular traits beyond the amyloid precursor protein. Aging (Albany NY) 2020; 12:1011-1033. [PMID: 31918411 PMCID: PMC6977673 DOI: 10.18632/aging.102677] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/25/2019] [Indexed: 02/07/2023]
Abstract
Alzheimer’s disease (AD) is the most prevalent type of dementia. Down syndrome (DS) is the leading genetic risk factor for Early-Onset AD, prematurely presenting the classic pathological features of the brain with AD. Augmented gene dosage, including the APP gene, could partially cause this predisposition. Recent works have revealed that alterations in chromosome location due to the extra Chromosome 21, as well as epigenetic modifications, could promote changes in gene expression other than those from Chromosome 21. As a result, similar pathological features and cellular dysfunctions in DS and AD, including impaired autophagy, lysosomal activity, and mitochondrial dysfunction, could be controlled beyond APP overexpression. In this review, we highlight some recent data regarding the origin of the shared features between DS and AD and explore the mechanisms concerning cognitive deficiencies in DS associated with dementia, which could shed some light into the search for new therapeutic targets for AD treatment.
Collapse
Affiliation(s)
- Wileidy Gomez
- Laboratory of Neuroprotection and Autophagy, Center for Integrative Biology, Faculty of Science, Universidad Mayor, Santiago, Chile.,Departamento de Bioquímica y Biología Molecular and Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Rodrigo Morales
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,CIBQA, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Vinicius Maracaja-Coutinho
- Departamento de Bioquímica y Biología Molecular and Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile.,Centro de Modelamiento Molecular, Biofísica y Bioinformática (CM2B2), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Valentina Parra
- Departamento de Bioquímica y Biología Molecular and Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile.,Center for Exercise, Metabolism, and Cancer Studies (CEMC), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Autophagy Research Center, Universidad de Chile, Santiago, Chile
| | - Melissa Nassif
- Laboratory of Neuroprotection and Autophagy, Center for Integrative Biology, Faculty of Science, Universidad Mayor, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| |
Collapse
|
23
|
Tramutola A, Lanzillotta C, Barone E, Arena A, Zuliani I, Mosca L, Blarzino C, Butterfield DA, Perluigi M, Di Domenico F. Intranasal rapamycin ameliorates Alzheimer-like cognitive decline in a mouse model of Down syndrome. Transl Neurodegener 2018; 7:28. [PMID: 30410750 PMCID: PMC6218962 DOI: 10.1186/s40035-018-0133-9] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/11/2018] [Indexed: 02/08/2023] Open
Abstract
Background Down syndrome (DS) individuals, by the age of 40s, are at increased risk to develop Alzheimer-like dementia, with deposition in brain of senile plaques and neurofibrillary tangles. Our laboratory recently demonstrated the disturbance of PI3K/AKT/mTOR axis in DS brain, prior and after the development of Alzheimer Disease (AD). The aberrant modulation of the mTOR signalling in DS and AD age-related cognitive decline affects crucial neuronal pathways, including insulin signaling and autophagy, involved in pathology onset and progression. Within this context, the therapeutic use of mTOR-inhibitors may prevent/attenuate the neurodegenerative phenomena. By our work we aimed to rescue mTOR signalling in DS mice by a novel rapamycin intranasal administration protocol (InRapa) that maximizes brain delivery and reduce systemic side effects. Methods Ts65Dn mice were administered with InRapa for 12 weeks, starting at 6 months of age demonstrating, at the end of the treatment by radial arms maze and novel object recognition testing, rescued cognition. Results The analysis of mTOR signalling, after InRapa, demonstrated in Ts65Dn mice hippocampus the inhibition of mTOR (reduced to physiological levels), which led, through the rescue of autophagy and insulin signalling, to reduced APP levels, APP processing and APP metabolites production, as well as, to reduced tau hyperphosphorylation. In addition, a reduction of oxidative stress markers was also observed. Discussion These findings demonstrate that chronic InRapa administration is able to exert a neuroprotective effect on Ts65Dn hippocampus by reducing AD pathological hallmarks and by restoring protein homeostasis, thus ultimately resulting in improved cognition. Results are discussed in term of a potential novel targeted therapeutic approach to reduce cognitive decline and AD-like neuropathology in DS individuals.
Collapse
Affiliation(s)
- Antonella Tramutola
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Chiara Lanzillotta
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Eugenio Barone
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.,2Universidad Autònoma de Chile, Instituto de Ciencias Biomédicas, Facultad de alud, Avenida Pedro de Valdivia 425, Providencia, Santiago, Chile
| | - Andrea Arena
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Ilaria Zuliani
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Luciana Mosca
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Carla Blarzino
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - D Allan Butterfield
- 3Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055 USA
| | - Marzia Perluigi
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Fabio Di Domenico
- 1Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
24
|
Calcineurin Silencing in Dictyostelium discoideum Leads to Cellular Alterations Affecting Mitochondria, Gene Expression, and Oxidative Stress Response. Protist 2018; 169:584-602. [DOI: 10.1016/j.protis.2018.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 04/04/2018] [Accepted: 04/11/2018] [Indexed: 11/18/2022]
|
25
|
Izzo A, Mollo N, Nitti M, Paladino S, Calì G, Genesio R, Bonfiglio F, Cicatiello R, Barbato M, Sarnataro V, Conti A, Nitsch L. Mitochondrial dysfunction in down syndrome: molecular mechanisms and therapeutic targets. Mol Med 2018; 24:2. [PMID: 30134785 PMCID: PMC6016872 DOI: 10.1186/s10020-018-0004-y] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 02/13/2018] [Indexed: 01/11/2023] Open
Abstract
Trisomy of chromosome 21 (TS21) is the most common autosomal aneuploidy compatible with postnatal survival with a prevalence of 1 in 700 newborns. Its phenotype is highly complex with constant features, such as mental retardation, dysmorphic traits and hypotonia, and variable features including heart defects, susceptibility to Alzheimer’s disease (AD), type 2 diabetes, obesity and immune disorders. Overexpression of genes on chromosome-21 (Hsa21) is responsible for the pathogenesis of Down syndrome (DS) phenotypic features either in a direct or in an indirect manner since many Hsa21 genes can affect the expression of other genes mapping to different chromosomes. Many of these genes are involved in mitochondrial function and energy conversion, and play a central role in the mitochondrial dysfunction and chronic oxidative stress, consistently observed in DS subjects. Recent studies highlight the deep interconnections between mitochondrial dysfunction and DS phenotype. In this short review we first provide a basic overview of mitochondrial phenotype in DS cells and tissues. We then discuss how specific Hsa21 genes may be involved in determining the disruption of mitochondrial DS phenotype and biogenesis. Finally we briefly focus on drugs that affect mitochondrial function and mitochondrial network suggesting possible therapeutic approaches to improve and/or prevent some aspects of the DS phenotype.
Collapse
Affiliation(s)
- Antonella Izzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Nunzia Mollo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Maria Nitti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Simona Paladino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Gaetano Calì
- Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - Rita Genesio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Ferdinando Bonfiglio
- Department of Biosciences and Nutrition, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Rita Cicatiello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Maria Barbato
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Viviana Sarnataro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| | - Anna Conti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy.
| | - Lucio Nitsch
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131, Naples, Italy
| |
Collapse
|
26
|
Di Domenico F, Tramutola A, Foppoli C, Head E, Perluigi M, Butterfield DA. mTOR in Down syndrome: Role in Aß and tau neuropathology and transition to Alzheimer disease-like dementia. Free Radic Biol Med 2018; 114:94-101. [PMID: 28807816 PMCID: PMC5748251 DOI: 10.1016/j.freeradbiomed.2017.08.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/07/2017] [Accepted: 08/09/2017] [Indexed: 12/12/2022]
Abstract
The mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase involved in the regulation of protein synthesis and degradation, longevity and cytoskeletal formation. The mTOR pathway represents a key growth and survival pathway involved in several diseases such as cancer, obesity, cardiovascular disease and neurodegenerative diseases. Numerous studies linked the alterations of mTOR pathway to age-dependent cognitive decline, pathogenesis of Alzheimer disease (AD) and AD-like dementia in Down syndrome (DS). DS is the most frequent chromosomal abnormality that causes intellectual disability. The neuropathology of AD in DS is complex and involves impaired mitochondrial function, defects in neurogenesis, increased oxidative stress, altered proteostasis and autophagy networks as a result of triplication of chromosome 21(chr 21). The chr21 gene products are considered a principal neuropathogenic moiety in DS. Several genes involved respectively in the formation of senile plaques and neurofibrillary tangles (NFT), two main pathological hallmarks of AD, are mapped on chr21. Further, in subjects with DS the activation of mTOR signaling contributes to Aβ generation and the formation of NFT. This review discusses recent research highlighting the complex role of mTOR associated with the presence of two hallmarks of AD pathology, senile plaques (composed mostly of fibrillar Aß peptides), and NFT (composed mostly of hyperphosphorylated tau protein). Oxidative stress, associated with chr21-related Aβ and mitochondrial alterations, may significantly contribute to this linkage of mTOR to AD-like neuropathology in DS.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Cesira Foppoli
- Department of Biochemical Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Elizabeth Head
- Sanders-Brown Center on Aging and Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506 USA.
| |
Collapse
|
27
|
Fu Q, Wu Y. RCAN1 in the inverse association between Alzheimer's disease and cancer. Oncotarget 2017; 9:54-66. [PMID: 29416595 PMCID: PMC5787488 DOI: 10.18632/oncotarget.23094] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/17/2017] [Indexed: 01/05/2023] Open
Abstract
The inverse association between Alzheimer’s disease (AD) and cancer has been reported in several population-based studies although both of them are age-related disorders. However, molecular mechanisms of the inverse association remain elusive. Increased expression of regulator of calcineurin 1 (RCAN1) promotes the pathogenesis of AD, while it suppresses cancer growth and progression in many types of cancer. Moreover, aberrant RCAN1 expression is detected in both AD and various types of cancer. It suggests that RCAN1 may play a key role in the inverse association between AD and cancer. In this article, we aim to review the role of RCAN1 in the inverse association and discuss underlying mechanisms, providing an insight into developing a novel approach to treat AD and cancer.
Collapse
Affiliation(s)
- Qiang Fu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yili Wu
- Department of Psychiatry, Jining Medical University, Jining, Shandong, China.,Shandong Key Laboratory of Behavioral Medicine, Jining, Shandong, China.,Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining, Shandong, China
| |
Collapse
|
28
|
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: 3.1] [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
|
29
|
Ballesteros-Martinez C, Mendez-Barbero N, Montalvo-Yuste A, Jensen BM, Gomez-Cardenosa A, Klitfod L, Garrido-Arandia M, Alvarez-Llamas G, Pastor-Vargas C, Vivanco F, Garvey LH, Cuesta-Herranz J, Poulsen LK, Esteban V. Endothelial Regulator of Calcineurin 1 Promotes Barrier Integrity and Modulates Histamine-Induced Barrier Dysfunction in Anaphylaxis. Front Immunol 2017; 8:1323. [PMID: 29104573 PMCID: PMC5655011 DOI: 10.3389/fimmu.2017.01323] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/29/2017] [Indexed: 12/21/2022] Open
Abstract
Anaphylaxis, the most serious and life-threatening allergic reaction, produces the release of inflammatory mediators by mast cells and basophils. Regulator of calcineurin 1 (Rcan1) is a negative regulator of mast-cell degranulation. The action of mediators leads to vasodilation and an increase in vascular permeability, causing great loss of intravascular volume in a short time. Nevertheless, the molecular basis remains unexplored on the vascular level. We investigated Rcan1 expression induced by histamine, platelet-activating factor (PAF), and epinephrine in primary human vein (HV)-/artery (HA)-derived endothelial cells (ECs) and human dermal microvascular ECs (HMVEC-D). Vascular permeability was analyzed in vitro in human ECs with forced Rcan1 expression using Transwell migration assays and in vivo using Rcan1 knockout mice. Histamine, but neither PAF nor epinephrine, induced Rcan1-4 mRNA and protein expression in primary HV-ECs, HA-ECs, and HMVEC-D through histamine receptor 1 (H1R). These effects were prevented by pharmacological inhibition of calcineurin with cyclosporine A. Moreover, intravenous histamine administration increased Rcan1 expression in lung tissues of mice undergoing experimental anaphylaxis. Functional in vitro assays showed that overexpression of Rcan1 promotes barrier integrity, suggesting a role played by this molecule in vascular permeability. Consistent with these findings, in vivo models of subcutaneous and intravenous histamine-mediated fluid extravasation showed increased response in skin, aorta, and lungs of Rcan1-deficient mice compared with wild-type animals. These findings reveal that endothelial Rcan1 is synthesized in response to histamine through a calcineurin-sensitive pathway and may reduce barrier breakdown, thus contributing to the strengthening of the endothelium and resistance to anaphylaxis. These new insights underscore its potential role as a regulator of sensitivity to anaphylaxis in humans.
Collapse
Affiliation(s)
| | - Nerea Mendez-Barbero
- Department of Vascular Physiopathology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Alma Montalvo-Yuste
- Department of Immunology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Bettina M Jensen
- Allergy Clinic, Gentofte Hospital, Copenhagen University Hospital, Hellerup, Denmark
| | | | - Lotte Klitfod
- Surgery Department, Gentofte Hospital, Copenhagen University Hospital, Hellerup, Denmark
| | - María Garrido-Arandia
- Center for Plant Biotechnology and Genomics, Technical University of Madrid, Madrid, Spain
| | - Gloria Alvarez-Llamas
- Department of Immunology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Carlos Pastor-Vargas
- Department of Immunology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Fernando Vivanco
- Department of Immunology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Lene Heise Garvey
- Allergy Clinic, Gentofte Hospital, Copenhagen University Hospital, Hellerup, Denmark
| | - Javier Cuesta-Herranz
- Department of Immunology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain.,Department of Allergy, Fundación Jiménez Díaz, Madrid, Spain
| | - Lars K Poulsen
- Allergy Clinic, Gentofte Hospital, Copenhagen University Hospital, Hellerup, Denmark
| | - Vanesa Esteban
- Department of Immunology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain.,Allergy Clinic, Gentofte Hospital, Copenhagen University Hospital, Hellerup, Denmark
| |
Collapse
|
30
|
Eckstein M, Vaeth M, Fornai C, Vinu M, Bromage TG, Nurbaeva MK, Sorge JL, Coelho PG, Idaghdour Y, Feske S, Lacruz RS. Store-operated Ca 2+ entry controls ameloblast cell function and enamel development. JCI Insight 2017; 2:e91166. [PMID: 28352661 DOI: 10.1172/jci.insight.91166] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Loss-of-function mutations in stromal interaction molecule 1 (STIM1) impair the activation of Ca2+ release-activated Ca2+ (CRAC) channels and store-operated Ca2+ entry (SOCE), resulting in a disease syndrome called CRAC channelopathy that is characterized by severe dental enamel defects. The cause of these enamel defects has remained unclear given a lack of animal models. We generated Stim1/2K14cre mice to delete STIM1 and its homolog STIM2 in enamel cells. These mice showed impaired SOCE in enamel cells. Enamel in Stim1/2K14cre mice was hypomineralized with decreased Ca content, mechanically weak, and thinner. The morphology of SOCE-deficient ameloblasts was altered, showing loss of the typical ruffled border, resulting in mislocalized mitochondria. Global gene expression analysis of SOCE-deficient ameloblasts revealed strong dysregulation of several pathways. ER stress genes associated with the unfolded protein response were increased in Stim1/2-deficient cells, whereas the expression of components of the glutathione system were decreased. Consistent with increased oxidative stress, we found increased ROS production, decreased mitochondrial function, and abnormal mitochondrial morphology in ameloblasts of Stim1/2K14cre mice. Collectively, these data show that loss of SOCE in enamel cells has substantial detrimental effects on gene expression, cell function, and the mineralization of dental enamel.
Collapse
Affiliation(s)
- Miriam Eckstein
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York, USA
| | - Martin Vaeth
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Cinzia Fornai
- Department of Anthropology, University of Vienna, Vienna, Austria.,Department of Evolutionary Medicine, University of Zurich, Zurich, Switzerland
| | - Manikandan Vinu
- Biology Program, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Timothy G Bromage
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York, USA.,Department of Biomaterials and Biomimetics, New York University College of Dentistry, New York, New York, USA
| | - Meerim K Nurbaeva
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York, USA
| | - Jessica L Sorge
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York, USA
| | - Paulo G Coelho
- Department of Biomaterials and Biomimetics, New York University College of Dentistry, New York, New York, USA
| | - Youssef Idaghdour
- Biology Program, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Rodrigo S Lacruz
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York, USA
| |
Collapse
|
31
|
Lim S, Hwang S, Yu JH, Lim JW, Kim H. Lycopene inhibits regulator of calcineurin 1-mediated apoptosis by reducing oxidative stress and down-regulating Nucling in neuronal cells. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201600530] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 11/16/2016] [Accepted: 11/24/2016] [Indexed: 02/04/2023]
Affiliation(s)
- Seiyoung Lim
- Department of Food and Nutrition; Brian Korea 21 PLUS Project; College of Human Ecology; Yonsei University; Seoul Republic of Korea
| | - Sinwoo Hwang
- Department of Food and Nutrition; Brian Korea 21 PLUS Project; College of Human Ecology; Yonsei University; Seoul Republic of Korea
| | - Ji Hoon Yu
- New Drug Development Center; Daegu-Gyeongbuk Medical Innovation Foundation; Daegu Korea
| | - Joo Weon Lim
- Department of Food and Nutrition; Brian Korea 21 PLUS Project; College of Human Ecology; Yonsei University; Seoul Republic of Korea
| | - Hyeyoung Kim
- Department of Food and Nutrition; Brian Korea 21 PLUS Project; College of Human Ecology; Yonsei University; Seoul Republic of Korea
| |
Collapse
|
32
|
|
33
|
Jiang H, Zhang C, Tang Y, Zhao J, Wang T, Liu H, Sun X. The regulator of calcineurin 1 increases adenine nucleotide translocator 1 and leads to mitochondrial dysfunctions. J Neurochem 2016; 140:307-319. [PMID: 27861892 PMCID: PMC5248620 DOI: 10.1111/jnc.13900] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 10/08/2016] [Accepted: 11/07/2016] [Indexed: 11/29/2022]
Abstract
The over‐expression of regulator of calcineurin 1 isoform 1 (RCAN1.1) has been implicated in mitochondrial dysfunctions of Alzheimer's disease; however, the mechanism linking RCAN1.1 over‐expression and the mitochondrial dysfunctions remains unknown. In this study, we use human neuroblastoma SH‐SY5Y cells stably expressing RCAN1.1S and rat primary neurons infected with RCAN1.1S expression lentivirus to study the association of RCAN1 with mitochondrial functions. Our study here showed that the over‐expression of RCAN1.1S remarkably up‐regulates the expression of adenine nucleotide translocator (ANT1) by stabilizing ANT1 mRNA. The increased ANT1 level leads to accelerated ATP–ADP exchange rate, more Ca2+‐induced mitochondrial permeability transition pore opening, increased cytochrome c release, and eventually cell apoptosis. Furthermore, knockdown of ANT1 expression brings these mitochondria perturbations caused by RCAN1.1S back to normal. The effect of RCAN1.1S on ANT1 was independent of its inhibition on calcineurin. This study elucidated a novel function of RCAN1 in mitochondria and provides a molecular basis for the RCAN1.1S over‐expression‐induced mitochondrial dysfunctions and neuronal apoptosis. ![]()
Collapse
Affiliation(s)
- Hui Jiang
- Otolaryngology Key Lab, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Department of Pediatrics, 2nd Hospital of Shandong University, Jinan, Shandong, China
| | - Chen Zhang
- Otolaryngology Key Lab, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yu Tang
- Otolaryngology Key Lab, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Juan Zhao
- Otolaryngology Key Lab, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Tan Wang
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Heng Liu
- Otolaryngology Key Lab, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiulian Sun
- Brain Research Institute, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
34
|
Wang G, Zhao Y, Liu S, Jia J, Lu T. Critical role of regulator of calcineurin 1 in spinal cord injury. J Physiol Biochem 2016; 72:605-613. [DOI: 10.1007/s13105-016-0499-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/14/2016] [Indexed: 10/21/2022]
|
35
|
Peiris H, Duffield MD, Fadista J, Jessup CF, Kashmir V, Genders AJ, McGee SL, Martin AM, Saiedi M, Morton N, Carter R, Cousin MA, Kokotos AC, Oskolkov N, Volkov P, Hough TA, Fisher EMC, Tybulewicz VLJ, Busciglio J, Coskun PE, Becker A, Belichenko PV, Mobley WC, Ryan MT, Chan JY, Laybutt DR, Coates PT, Yang S, Ling C, Groop L, Pritchard MA, Keating DJ. A Syntenic Cross Species Aneuploidy Genetic Screen Links RCAN1 Expression to β-Cell Mitochondrial Dysfunction in Type 2 Diabetes. PLoS Genet 2016; 12:e1006033. [PMID: 27195491 PMCID: PMC4873152 DOI: 10.1371/journal.pgen.1006033] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 04/18/2016] [Indexed: 12/20/2022] Open
Abstract
Type 2 diabetes (T2D) is a complex metabolic disease associated with obesity, insulin resistance and hypoinsulinemia due to pancreatic β-cell dysfunction. Reduced mitochondrial function is thought to be central to β-cell dysfunction. Mitochondrial dysfunction and reduced insulin secretion are also observed in β-cells of humans with the most common human genetic disorder, Down syndrome (DS, Trisomy 21). To identify regions of chromosome 21 that may be associated with perturbed glucose homeostasis we profiled the glycaemic status of different DS mouse models. The Ts65Dn and Dp16 DS mouse lines were hyperglycemic, while Tc1 and Ts1Rhr mice were not, providing us with a region of chromosome 21 containing genes that cause hyperglycemia. We then examined whether any of these genes were upregulated in a set of ~5,000 gene expression changes we had identified in a large gene expression analysis of human T2D β-cells. This approach produced a single gene, RCAN1, as a candidate gene linking hyperglycemia and functional changes in T2D β-cells. Further investigations demonstrated that RCAN1 methylation is reduced in human T2D islets at multiple sites, correlating with increased expression. RCAN1 protein expression was also increased in db/db mouse islets and in human and mouse islets exposed to high glucose. Mice overexpressing RCAN1 had reduced in vivo glucose-stimulated insulin secretion and their β-cells displayed mitochondrial dysfunction including hyperpolarised membrane potential, reduced oxidative phosphorylation and low ATP production. This lack of β-cell ATP had functional consequences by negatively affecting both glucose-stimulated membrane depolarisation and ATP-dependent insulin granule exocytosis. Thus, from amongst the myriad of gene expression changes occurring in T2D β-cells where we had little knowledge of which changes cause β-cell dysfunction, we applied a trisomy 21 screening approach which linked RCAN1 to β-cell mitochondrial dysfunction in T2D.
Collapse
Affiliation(s)
- Heshan Peiris
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Michael D. Duffield
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | | | - Claire F. Jessup
- Islet Biology Laboratory, Department of Anatomy and Histology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Vinder Kashmir
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Amanda J. Genders
- Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | - Sean L. McGee
- Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
- Metabolism and Inflammation Program, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Alyce M. Martin
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Madiha Saiedi
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Nicholas Morton
- Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Roderick Carter
- Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael A. Cousin
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Alexandros C. Kokotos
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Petr Volkov
- Lund University Diabetes Centre, Malmö, Sweden
| | - Tertius A. Hough
- Mary Lyon Centre Pathology, MRC Harwell, Harwell Oxford Science Park, Oxford, United Kingdom
| | - Elizabeth M. C. Fisher
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Victor L. J. Tybulewicz
- Francis Crick Institute, Mill Hill, London, United Kingdom
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Jorge Busciglio
- Department of Neurobiology and Behaviour, University of California, Irvine, Irvine, California, United States of America
| | - Pinar E. Coskun
- Department of Neurobiology and Behaviour, University of California, Irvine, Irvine, California, United States of America
| | - Ann Becker
- Department of Neurosciences School of Medicine, University of California, San Diego, San Diego, California, United States of America
| | - Pavel V. Belichenko
- Department of Neurosciences School of Medicine, University of California, San Diego, San Diego, California, United States of America
| | - William C. Mobley
- Department of Neurosciences School of Medicine, University of California, San Diego, San Diego, California, United States of America
| | - Michael T. Ryan
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - Jeng Yie Chan
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - D. Ross Laybutt
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - P. Toby Coates
- Clinical and Experimental Transplantation Group, Royal Adelaide Hospital, North Terrace, Adelaide, South Australia, Australia
| | - Sijun Yang
- Animal Experiment Center, Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China
| | | | - Leif Groop
- Lund University Diabetes Centre, Malmö, Sweden
| | - Melanie A. Pritchard
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - Damien J. Keating
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| |
Collapse
|
36
|
Effects of sarah/nebula knockdown on Aβ42-induced phenotypes during Drosophila development. Genes Genomics 2016. [DOI: 10.1007/s13258-016-0407-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
37
|
Lee S, Bang SM, Hong YK, Lee JH, Jeong H, Park SH, Liu QF, Lee IS, Cho KS. The calcineurin inhibitor Sarah (Nebula) exacerbates Aβ42 phenotypes in a Drosophila model of Alzheimer's disease. Dis Model Mech 2015; 9:295-306. [PMID: 26659252 PMCID: PMC4826976 DOI: 10.1242/dmm.018069] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 11/28/2015] [Indexed: 11/20/2022] Open
Abstract
Expression of the Down syndrome critical region 1 (DSCR1) protein, an inhibitor of the Ca2+-dependent phosphatase calcineurin, is elevated in the brains of individuals with Down syndrome (DS) or Alzheimer's disease (AD). Although increased levels of DSCR1 were often observed to be deleterious to neuronal health, its beneficial effects against AD neuropathology have also been reported, and the roles of DSCR1 on the pathogenesis of AD remain controversial. Here, we investigated the role of sarah (sra; also known as nebula), a Drosophila DSCR1 ortholog, in amyloid-β42 (Aβ42)-induced neurological phenotypes in Drosophila. We detected sra expression in the mushroom bodies of the fly brain, which are a center for learning and memory in flies. Moreover, similar to humans with AD, Aβ42-expressing flies showed increased Sra levels in the brain, demonstrating that the expression pattern of DSCR1 with regard to AD pathogenesis is conserved in Drosophila. Interestingly, overexpression of sra using the UAS-GAL4 system exacerbated the rough-eye phenotype, decreased survival rates and increased neuronal cell death in Aβ42-expressing flies, without modulating Aβ42 expression. Moreover, neuronal overexpression of sra in combination with Aβ42 dramatically reduced both locomotor activity and the adult lifespan of flies, whereas flies with overexpression of sra alone showed normal climbing ability, albeit with a slightly reduced lifespan. Similarly, treatment with chemical inhibitors of calcineurin, such as FK506 and cyclosporin A, or knockdown of calcineurin expression by RNA interference (RNAi), exacerbated the Aβ42-induced rough-eye phenotype. Furthermore, sra-overexpressing flies displayed significantly decreased mitochondrial DNA and ATP levels, as well as increased susceptibility to oxidative stress compared to that of control flies. Taken together, our results demonstrating that sra overexpression augments Aβ42 cytotoxicity in Drosophila suggest that DSCR1 upregulation or calcineurin downregulation in the brain might exacerbate Aβ42-associated neuropathogenesis in AD or DS. Drosophila Collection: Chronically increased levels of Sarah (Nebula), a calcineurin inhibitor, cause mitochondria dysfunction and subsequently increased Aβ42-induced cytotoxicity in Drosophila.
Collapse
Affiliation(s)
- Soojin Lee
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Se Min Bang
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Yoon Ki Hong
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Jang Ho Lee
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Haemin Jeong
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Seung Hwan Park
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Quan Feng Liu
- Department of Oriental Medicine, Dongguk University, Gyeogju 38066, Republic of Korea Department of Oriental Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, Gyeonggi 10326, Republic of Korea
| | - Im-Soon Lee
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyoung Sang Cho
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
38
|
Wong H, Levenga J, Cain P, Rothermel B, Klann E, Hoeffer C. RCAN1 overexpression promotes age-dependent mitochondrial dysregulation related to neurodegeneration in Alzheimer's disease. Acta Neuropathol 2015; 130:829-43. [PMID: 26497675 DOI: 10.1007/s00401-015-1499-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 10/15/2015] [Accepted: 10/16/2015] [Indexed: 10/22/2022]
Abstract
Aging is the largest risk factor for Alzheimer's disease (AD). Patients with Down syndrome (DS) develop symptoms consistent with early-onset AD, suggesting that overexpression of chromosome 21 genes such as Regulator of Calcineurin 1 (RCAN1) plays a role in AD pathogenesis. RCAN1 levels are increased in the brain of DS and AD patients but also in the human brain with normal aging. RCAN1 has been implicated in several neuronal functions, but whether its increased expression is correlative or causal in the aging-related progression of AD remains elusive. We show that brain-specific overexpression of the human RCAN1.1S isoform in mice promotes early age-dependent memory and synaptic plasticity deficits, tau pathology, and dysregulation of dynamin-related protein 1 (DRP1) activity associated with mitochondrial dysfunction and oxidative stress, reproducing key AD features. Based on these findings, we propose that chronic RCAN1 overexpression during aging alters DRP1-mediated mitochondrial fission and thus acts to promote AD-related progressive neurodegeneration.
Collapse
Affiliation(s)
- Helen Wong
- Center for Neural Science, New York University, New York, NY, USA
| | - Josien Levenga
- Department of Integrated of Physiology, Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
| | - Peter Cain
- Department of Integrated of Physiology, Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
| | - Beverly Rothermel
- Department of Cardiology, University of Texas-Southwestern, Dallas, TX, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY, USA
| | - Charles Hoeffer
- Department of Integrated of Physiology, Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA.
- New York University School of Medicine, New York, NY, USA.
- Linda Crnic Institute, Denver, CO, USA.
| |
Collapse
|
39
|
Xu W, Barrientos T, Mao L, Rockman HA, Sauve AA, Andrews NC. Lethal Cardiomyopathy in Mice Lacking Transferrin Receptor in the Heart. Cell Rep 2015; 13:533-545. [PMID: 26456827 DOI: 10.1016/j.celrep.2015.09.023] [Citation(s) in RCA: 216] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 08/21/2015] [Accepted: 09/04/2015] [Indexed: 01/26/2023] Open
Abstract
Both iron overload and iron deficiency have been associated with cardiomyopathy and heart failure, but cardiac iron utilization is incompletely understood. We hypothesized that the transferrin receptor (Tfr1) might play a role in cardiac iron uptake and used gene targeting to examine the role of Tfr1 in vivo. Surprisingly, we found that decreased iron, due to inactivation of Tfr1, was associated with severe cardiac consequences. Mice lacking Tfr1 in the heart died in the second week of life and had cardiomegaly, poor cardiac function, failure of mitochondrial respiration, and ineffective mitophagy. The phenotype could only be rescued by aggressive iron therapy, but it was ameliorated by administration of nicotinamide riboside, an NAD precursor. Our findings underscore the importance of both Tfr1 and iron in the heart, and may inform therapy for patients with heart failure.
Collapse
Affiliation(s)
- Wenjing Xu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27705, USA
| | - Tomasa Barrientos
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27705, USA
| | - Lan Mao
- Department of Medicine, Duke University School of Medicine, Duke University, Durham, NC 27705, USA
| | - Howard A Rockman
- Department of Medicine, Duke University School of Medicine, Duke University, Durham, NC 27705, USA
| | - Anthony A Sauve
- Department of Pharmacology, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Nancy C Andrews
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Duke University, Durham, NC 27705, USA; Department of Pediatrics, Duke University School of Medicine, Duke University, Durham, NC 27705, USA.
| |
Collapse
|
40
|
Mitochondrial degradation and energy metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2812-21. [DOI: 10.1016/j.bbamcr.2015.05.010] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 04/23/2015] [Accepted: 05/07/2015] [Indexed: 12/14/2022]
|
41
|
Lee EH, Kim SS, Lee S, Baek KH, Seo SR. Pituitary Adenylate Cyclase-activating Polypeptide (PACAP) Targets Down Syndrome Candidate Region 1 (DSCR1/RCAN1) to control Neuronal Differentiation. J Biol Chem 2015; 290:21019-21031. [PMID: 26157140 DOI: 10.1074/jbc.m115.639476] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Indexed: 11/06/2022] Open
Abstract
Pituitary adenylate cyclase-activating peptide (PACAP) is a neurotrophic peptide involved in a wide range of nervous functions, including development, differentiation, and survival, and various aspects of learning and memory. Here we report that PACAP induces the expression of regulator of calcineurin 1 (RCAN1, also known as DSCR1), which is abnormally expressed in the brains of Down syndrome patients. Increased RCAN1 expression is accompanied by activation of the PKA-cAMP response element-binding protein pathways. EMSA and ChIP analyses demonstrate the presence of a functional cAMP response element in the RCAN1 promoter. Moreover, we show that PACAP-dependent neuronal differentiation is significantly disturbed by improper RCAN1 expression. Our data provide the first evidence of RCAN1, a Down syndrome-related gene, as a novel target for control of the neurotrophic function of PACAP.
Collapse
Affiliation(s)
- Eun Hye Lee
- Department of Molecular Bioscience, College of Biomedical Science, and Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 200-701, Republic of Korea and
| | - Seon Sook Kim
- Department of Molecular Bioscience, College of Biomedical Science, and Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 200-701, Republic of Korea and
| | - Seul Lee
- Department of Molecular and Cellular Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi 440-746, Republic of Korea
| | - Kwan-Hyuck Baek
- Department of Molecular and Cellular Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi 440-746, Republic of Korea
| | - Su Ryeon Seo
- Department of Molecular Bioscience, College of Biomedical Science, and Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 200-701, Republic of Korea and.
| |
Collapse
|
42
|
Lloret A, Fuchsberger T, Giraldo E, Viña J. Molecular mechanisms linking amyloid β toxicity and Tau hyperphosphorylation in Alzheimer׳s disease. Free Radic Biol Med 2015; 83:186-91. [PMID: 25746773 DOI: 10.1016/j.freeradbiomed.2015.02.028] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 12/12/2022]
Abstract
Neurofibrillary tangles (aggregates of cytoskeletal Tau protein) and senile plaques (aggregates mainly formed by amyloid β peptide) are two landmark lesions in Alzheimer׳s disease. Some researchers have proposed tangles, whereas others have proposed plaques, as primary lesions. For a long time, these were thought of as independent mechanisms. However, experimental evidence suggests that both lesions are intimately related. We review here some molecular pathways linking amyloid β and Tau toxicities involving, among others, glycogen synthase kinase 3β, p38, Pin1, cyclin-dependent kinase 5, and regulator of calcineurin 1. Understanding amyloid β and Tau toxicities as part of a common pathophysiological mechanism may help to find molecular targets to prevent or even treat the disease.
Collapse
Affiliation(s)
- A Lloret
- Department of Physiology, Faculty of Medicine, University of Valencia and Fundacion Investigacion Hospital Clinico Universitario/INCLIVA, 46010 Valencia, Spain
| | - T Fuchsberger
- Department of Physiology, Faculty of Medicine, University of Valencia and Fundacion Investigacion Hospital Clinico Universitario/INCLIVA, 46010 Valencia, Spain
| | - E Giraldo
- Department of Physiology, Faculty of Medicine, University of Valencia and Fundacion Investigacion Hospital Clinico Universitario/INCLIVA, 46010 Valencia, Spain
| | - J Viña
- Department of Physiology, Faculty of Medicine, University of Valencia and Fundacion Investigacion Hospital Clinico Universitario/INCLIVA, 46010 Valencia, Spain.
| |
Collapse
|
43
|
Abstract
The metabolism of malignant cells is profoundly altered in order to maintain their survival and proliferation in adverse microenvironmental conditions. Autophagy is an intracellular recycling process that maintains basal levels of metabolites and biosynthetic intermediates under starvation or other forms of stress, hence serving as an important mechanism for metabolic adaptation in cancer cells. Although it is widely acknowledged that autophagy sustains metabolism in neoplastic cells under duress, many questions remain with regard to the mutual relationship between autophagy and metabolism in cancer. Importantly, autophagy has often been described as a "double-edged sword" that can either impede or promote cancer initiation and progression. Here, we overview such a dual function of autophagy in tumorigenesis and our current understanding of the coordinated regulation of autophagy and cancer cell metabolism in the control of tumor growth, progression, and resistance to therapy.
Collapse
|
44
|
Duan H, Li Y, Yan L, Yang H, Wu J, Qian P, Li B, Wang S. Rcan1-1L overexpression induces mitochondrial autophagy and improves cell survival in angiotensin II-exposed cardiomyocytes. Exp Cell Res 2015; 335:99-106. [PMID: 25978972 DOI: 10.1016/j.yexcr.2015.05.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 04/15/2015] [Accepted: 05/04/2015] [Indexed: 01/09/2023]
Abstract
Mitochondrial autophagy is an important adaptive stress response and can be modulated by various key molecules. A previous study found that the regulator of calcineurin 1-1L (Rcan1-1L) may regulate mitochondrial autophagy and cause mitochondria degradation in neurocytes. However, the effect of Rcan1-1L on cardiomyocytes has not been determined. In the present study, we aimed to investigate the role of Rcan1-1L in angiotensin II (Ang II)-exposed human cardiomyocytes. Above all, Human adult cardiac myocytes (HACMs) were exposed to 200nmol/L Ang II for 4 days. Enhanced H2O2 production, cytochrome C release and mitochondrial permeability were observed in these cells, which were blocked by valsartan. Consistently, Ang II exposure significantly reduced cardiomyocyte viability. However, transfection of Rcan1-1L vector promoted cell viability and ameliorated the apoptosis caused by Ang II. Rcan1-1L clearly promoted mitochondrial autophagy in HACMs, with elevated autophagy protein (ATG) 5 and light chain 3 (LC3) expression. Transient mitochondrial biogenesis and reduced cytochrome C release was also induced by Rcan1-1L. Additionally, Rcan1-1L significantly inhibited calcineurin/nuclear factor of activated T cells (NFAT) signaling. We thus conclude that Rcan1-1L may play a protective role in Ang II-treated cardiomyocytes through the induction of mitochondrial autophagy, and may be an alternative method of cardiac protection.
Collapse
Affiliation(s)
- Hongyan Duan
- Department of cardiology, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Yongqiang Li
- Department of cardiology, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Lijie Yan
- Department of cardiology, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Haitao Yang
- Department of cardiology, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Jintao Wu
- Department of cardiology, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Peng Qian
- Department of cardiology, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Bing Li
- Department of cardiology, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Shanling Wang
- Department of cardiology, Henan Provincial People's Hospital, Zhengzhou 450003, PR China.
| |
Collapse
|
45
|
Wu Y, Deng Y, Zhang S, Luo Y, Cai F, Zhang Z, Zhou W, Li T, Song W. Amyloid-β precursor protein facilitates the regulator of calcineurin 1-mediated apoptosis by downregulating proteasome subunit α type-5 and proteasome subunit β type-7. Neurobiol Aging 2015; 36:169-177. [PMID: 25194880 DOI: 10.1016/j.neurobiolaging.2014.07.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/23/2014] [Accepted: 07/23/2014] [Indexed: 11/28/2022]
Abstract
Individuals with Down syndrome (DS), caused by trisomy of chromosome 21, inevitably develop characteristic Alzheimer's disease (AD) neuropathology, including neuritic plaques, neurofibrillary tangles, and neuronal loss. Amyloid-β protein, the major component of neuritic plaques, is the proteolytic product of amyloid-β precursor protein (APP). APP and the regulator of calcineurin 1 (RCAN1) genes on chromosome 21 play a pivotal role in promoting plaque formation and neuronal apoptosis. However, the mechanism underlying AD pathogenesis in DS is not well defined. In this study, we demonstrated that APP significantly increased RCAN1 level in both cells and transgenic mice. Overexpression of APP significantly reduced the expression of 2 proteasome subunits, proteasome subunit α type-5 and proteasome subunit β type-7, leading to the inhibition of proteasomal degradation of RCAN1. Furthermore, knockdown of RCAN1 expression attenuated APP-induced neuronal apoptosis. Taken together, the results clearly showed that APP has a previously unknown function in regulating RCAN1-mediated neuronal apoptosis through the proteasome pathway. Our study demonstrates a novel mechanism by which overexpression of APP and RCAN1 causes neurodegeneration and AD pathogenesis in DS, and it provides new insights into the potential of targeting APP-induced proteasomal impairment and RCAN1 accumulation for AD and DS treatment.
Collapse
Affiliation(s)
- Yili Wu
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders and Ministry of Education Key Lab of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Yu Deng
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Shuting Zhang
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Yawen Luo
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Fang Cai
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Zhuohua Zhang
- State Key Laboratory of Medical Genetics of China, Central South University, Changsha, Hunan, China
| | - Weihui Zhou
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders and Ministry of Education Key Lab of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Tingyu Li
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders and Ministry of Education Key Lab of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Weihong Song
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders and Ministry of Education Key Lab of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
46
|
Sun L, Hao Y, An R, Li H, Xi C, Shen G. Overexpression of Rcan1-1L inhibits hypoxia-induced cell apoptosis through induction of mitophagy. Mol Cells 2014; 37:785-94. [PMID: 25377251 PMCID: PMC4255098 DOI: 10.14348/molcells.2014.0103] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 09/10/2014] [Accepted: 09/11/2014] [Indexed: 12/21/2022] Open
Abstract
Mitophagy, a cellular process that selectively targets dysfunctional mitochondria for degradation, is currently a hot topic in research into the pathogenesis and treatment of many human diseases. Considering that hypoxia causes mitochondrial dysfunction, which results in cell death, we speculated that selective activation of mitophagy might promote cell survival under hypoxic conditions. In the present study, we introduced the Regulator of calcineurin 1-1L (Rcan1-1L) to initiate the mitophagy pathway and aimed to evaluate the effect of Rcan1-1L-induced mitophagy on cell survival under hypoxic conditions. Recombinant adenovirus vectors carrying Rcan1-1L were transfected into human umbilical vein endothelial cells and human adult cardiac myocytes. Using the 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide MTT assay and Trypan blue exclusion assay, Rcan1-1L overexpression was found to markedly reverse cell growth inhibition induced by hypoxia. Additionally, Rcan1-1L overexpression inhibited cell apoptosis under hypoxic conditions, as detected by annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) apoptosis assay. Meanwhile, the mitochondria-mediated cell apoptotic pathway was inhibited by Rcan1-1L. In contrast, knockdown of Rcan1-1L accelerated hypoxia-induced cell apoptosis. Moreover, Rcan1-1L overexpression significantly reduced mitochondrial mass, decreased depolarized mitochondria, and downregulated ATP and reactive oxygen species production. We further delineated that the loss of mitochondrial mass was due to the activation of mitophagy induced by Rcan1-1L. Rcan1-1L overexpression activated autophagy flux and promoted translocation of the specific mitophagy receptor Parkin into mitochondria from the cytosol, whereas inhibition of autophagy flux resulted in the accumulation of Parkin-loaded mitochondria. Finally, we demonstrated that mitochondrial permeability transition pore opening was significantly increased by Rcan1-1L overexpression, which suggested that Rcan1-1L might evoke mitophagy through regulating mitochondrial permeability transition pores. Taken together, we provide evidence that Rcan1-1L overexpression induces mitophagy, which in turn contributes to cell survival under hypoxic conditions, revealing for the first time that Rcan1-1L-induced mitophagy may be used for cardioprotection.
Collapse
Affiliation(s)
- Lijun Sun
- Department of Radiology, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi 710032,
P.R. China
| | - Yuewen Hao
- Department of Radiology, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi 710032,
P.R. China
| | - Rui An
- Department of Radiology, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi 710032,
P.R. China
| | - Haixun Li
- Department of Radiology, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi 710032,
P.R. China
| | - Cong Xi
- Department of Radiology, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi 710032,
P.R. China
| | - Guohong Shen
- Department of Radiology, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi 710032,
P.R. China
| |
Collapse
|
47
|
Pi H, Xu S, Zhang L, Guo P, Li Y, Xie J, Tian L, He M, Lu Y, Li M, Zhang Y, Zhong M, Xiang Y, Deng L, Zhou Z, Yu Z. Dynamin 1-like-dependent mitochondrial fission initiates overactive mitophagy in the hepatotoxicity of cadmium. Autophagy 2014; 9:1780-800. [DOI: 10.4161/auto.25665] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
48
|
Regulator of Calcineurin 1-1L Protects Cardiomyocytes Against Hypoxia-induced Apoptosis via Mitophagy. J Cardiovasc Pharmacol 2014; 64:310-7. [DOI: 10.1097/fjc.0000000000000121] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
49
|
GONG GU, HU LING, LIU YINGHAI, BAI SHURONG, DAI XUEMEI, YIN LIANG, SUN YANGYANG, WANG XIAOWU, HOU LICHAO. Upregulation of HIF-1α protein induces mitochondrial autophagy in primary cortical cell cultures through the inhibition of the mTOR pathway. Int J Mol Med 2014; 34:1133-40. [DOI: 10.3892/ijmm.2014.1850] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 06/26/2014] [Indexed: 11/06/2022] Open
|
50
|
RCAN1 regulates mitochondrial function and increases susceptibility to oxidative stress in mammalian cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:520316. [PMID: 25009690 PMCID: PMC4070399 DOI: 10.1155/2014/520316] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 05/06/2014] [Indexed: 12/02/2022]
Abstract
Mitochondria are the primary site of cellular energy generation and reactive oxygen species (ROS) accumulation. Elevated ROS levels are detrimental to normal cell function and have been linked to the pathogenesis of neurodegenerative disorders such as Down's syndrome (DS) and Alzheimer's disease (AD). RCAN1 is abundantly expressed in the brain and overexpressed in brain of DS and AD patients. Data from nonmammalian species indicates that increased RCAN1 expression results in altered mitochondrial function and that RCAN1 may itself regulate neuronal ROS production. In this study, we have utilized mice overexpressing RCAN1 (RCAN1ox) and demonstrate an increased susceptibility of neurons from these mice to oxidative stress. Mitochondria from these mice are more numerous and smaller, indicative of mitochondrial dysfunction, and mitochondrial membrane potential is altered under conditions of oxidative stress. We also generated a PC12 cell line overexpressing RCAN1 (PC12RCAN1). Similar to RCAN1ox neurons, PC12RCAN1 cells have an increased susceptibility to oxidative stress and produce more mitochondrial ROS. This study demonstrates that increasing RCAN1 expression alters mitochondrial function and increases the susceptibility of neurons to oxidative stress in mammalian cells. These findings further contribute to our understanding of RCAN1 and its potential role in the pathogenesis of neurodegenerative disorders such as AD and DS.
Collapse
|