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
|
Yang K, Li T, Geng Y, Zou X, Peng F, Gao W. The role of mitophagy in the development of chronic kidney disease. PeerJ 2024; 12:e17260. [PMID: 38680884 PMCID: PMC11056108 DOI: 10.7717/peerj.17260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 03/28/2024] [Indexed: 05/01/2024] Open
Abstract
Chronic kidney disease (CKD) represents a significant global health concern, with renal fibrosis emerging as a prevalent and ultimate manifestation of this condition. The absence of targeted therapies presents an ongoing and substantial challenge. Accumulating evidence suggests that the integrity and functionality of mitochondria within renal tubular epithelial cells (RTECs) often become compromised during CKD development, playing a pivotal role in the progression of renal fibrosis. Mitophagy, a specific form of autophagy, assumes responsibility for eliminating damaged mitochondria to uphold mitochondrial equilibrium. Dysregulated mitophagy not only correlates with disrupted mitochondrial dynamics but also contributes to the advancement of renal fibrosis in CKD. While numerous studies have examined mitochondrial metabolism, ROS (reactive oxygen species) production, inflammation, and apoptosis in kidney diseases, the precise pathogenic mechanisms underlying mitophagy in CKD remain elusive. The exact mechanisms through which modulating mitophagy mitigates renal fibrosis, as well as its influence on CKD progression and prognosis, have not undergone systematic investigation. The role of mitophagy in AKI has been relatively clear, but the role of mitophagy in CKD is still rare. This article presents a comprehensive review of the current state of research on regulating mitophagy as a potential treatment for CKD. The objective is to provide fresh perspectives, viable strategies, and practical insights into CKD therapy, thereby contributing to the enhancement of human living conditions and patient well-being.
Collapse
Affiliation(s)
- Kexin Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Ting Li
- Department of Pathophysiology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Yingpu Geng
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Xiangyu Zou
- Department of Pathophysiology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Fujun Peng
- Department of Pathophysiology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Wei Gao
- Department of Pathophysiology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| |
Collapse
|
3
|
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
|
4
|
Zhang J, Chen H, Weng X, Liu H, Chen Z, Huang Q, Wang L, Liu X. RCAN1.4 attenuates renal fibrosis through inhibiting calcineurin-mediated nuclear translocation of NFAT2. Cell Death Discov 2021; 7:317. [PMID: 34707090 PMCID: PMC8551295 DOI: 10.1038/s41420-021-00713-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/21/2021] [Accepted: 10/13/2021] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is thus deemed to a global health problem. Renal fibrosis, characterized by accumulation of extracellular matrix (ECM) components in the kidney, is considered a common pathway leading to CKD. Regulator of calcineurin1 (RCAN1), identified as a competitive endogenous inhibitor of the phosphatase calcineurin, participates in ECM deposition in various organs. However, the role of RCAN1 in renal fibrosis remains unclear. Here, unilateral ureteral obstruction (UUO), a well-known model to induce renal fibrosis in vivo, was performed on mice for a week. To overexpress RCAN1.4 in vivo, recombinant adeno-associated virus 9-packed RCAN1.4 over-expression plasm was employed in mice kidney. Lentivirus-packed RCAN1.4 over-expression plasm was employed to transfer into HK-2 and NRK-49F cells in vitro. The results indicated that RCAN1.4 expression was impaired both in UUO-induced renal fibrosis in vivo and TGF-β1-induced renal fibrosis in vitro. However, knocking in of RCAN1.4 suppressed the production of extracellular matrix (ECM) both in vivo and in vitro. Furthermore, in vitro, the apoptosis-related proteins, including the ratio of Bax/Bcl-2 and cleaved-caspase3, were elevated in cells transfected with RCAN1.4 overexpression plasmid. In addition, we found that RCAN1.4 could rugulated NFAT2 nuclear distribution by inhibiting calcineurin pathway. So overexpression of RCAN1.4 could reverse renal fibrosis, attenuate ECM related protein accumulation, promote apoptosis of myofibroblast via inhibiting Calcineurin/NFAT2 signaling pathway. Taken together, our study demonstrated that targeting RCAN1.4 may be therapeutic efficacy in renal fibrosis.
Collapse
Affiliation(s)
- Jianjian Zhang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Hui Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xiaodong Weng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Hao Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Qin Huang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 430060, Wuhan, Hubei, China.
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
5
|
Wang Y, Wang Q, Chen J, Yao LH, Tang N, Jiang ZX, Luo Y. Protective Effect of Hydroxysafflor Yellow A against Chronic Mild Stress-induced Memory Impairments by Suppressing Tau Phosphorylation in Mice. Curr Med Sci 2021; 41:555-564. [PMID: 34129201 DOI: 10.1007/s11596-021-2369-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 09/10/2020] [Indexed: 12/19/2022]
Abstract
Chronic stress plays a critical role in the etiology of sporadic Alzheimer's disease (AD). However, there are currently no effective drugs that can target chronic stress to prevent AD. In this study, we explored the neuroprotective effect of hydroxysafflor yellow A (HSYA) against chronic mild stress (CMS)-induced memory impairments in mice and the underlying mechanism. The Morris water maze test showed that HSYA significantly reduced CMS-induced learning and memory impairments in mice. HSYA increased the expression of brain-derived neurotrophic factor (BDNF) and activated downstream tropomyosin-related kinase B (TrkB) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B(Akt)/mammalian target of rapamycin (mTOR) signaling. HSYA decreased the expression of regulator of calcineurin 1-1L (RCAN1-1L) that could promote the activity of glycogen synthase kinase-3β (GSK-3β). HSYA also attenuated tau phosphorylation by inhibiting the activity of GSK-3β and cyclin-dependent kinase-5 (Cdk5). Our data indicated that HSYA has protective effects against CMS-induced BDNF downregulation, tau phosphorylation and memory impairments. HSYA may be a promising therapeutic candidate for AD by targeting chronic stress.
Collapse
Affiliation(s)
- Ying Wang
- Department of Neurology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Qiang Wang
- Department of Neurology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Jun Chen
- Department of Neurology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Li-He Yao
- Department of Neurology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Ni Tang
- Department of Neurology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Zhen-Xiu Jiang
- Department of Neurology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Yu Luo
- Institute of Pathophysiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
6
|
Identification of new BACE1 inhibitors for treating Alzheimer's disease. J Mol Model 2021; 27:58. [PMID: 33517514 DOI: 10.1007/s00894-021-04679-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 01/14/2021] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is a type of brain disorder, wherein a person experiences gradual memory loss, state of confusion, hallucination, agitation, and personality change. AD is marked by the presence of extracellular amyloid plaques and intracellular neurofibrillary tangles (NFTs) and synaptic losses. Increased cases of AD in recent times created a dire need to discover or identify chemical compounds that can cease the development of AD. This study focuses on finding potential drug molecule(s) active against β-secretase, also known as β-site amyloid precursor protein cleaving enzyme 1 (BACE1). Clustering analysis followed by phylogenetic studies on microarray datasets retrieved from GEO browser showed that BACE1 gene has genetic relatedness with the RCAN1 gene. A ligand library comprising 60 natural compounds retrieved from literature and 25 synthetic compounds collected from DrugBank were screened. Further, 350 analogues of potential parent compounds were added to the library for the docking purposes. Molecular docking studies identified 11-oxotigogenin as the best ligand molecule. The compound showed the binding affinity of - 11.1 Kcal/mole and forms three hydrogen bonds with Trp124, Ile174, and Arg176. The protein-ligand complex was subjected to 25 ns molecular dynamics simulation and the potential energy of the complex was found to be - 1.24579e+06 Kcal/mole. In this study, 11-oxotigogenin has shown promising results against BACE1, which is a leading cause of AD, hence warrants for in vitro and in vivo validation of the same. In addition, in silico identification of 11-oxotigogenin as a potential anti-AD compound paves the way for designing of chemical scaffolds to discover more potent BACE1 inhibitors.Graphical abstract.
Collapse
|
7
|
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
|
8
|
Sang XY, Xiao JJ, Liu Q, Zhu R, Dai JJ, Zhang C, Yu H, Yang SJ, Zhang BF. Regulators of calcineurin 1 deficiency attenuates tubulointerstitial fibrosis through improving mitochondrial fitness. FASEB J 2020; 34. [PMID: 32896034 DOI: 10.1096/fj.202000781rrr] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023]
Abstract
Renal fibrosis is the common pathological process of various chronic kidney diseases (CKD). Recent studies indicate that mitochondrial fragmentation is closely associated with renal fibrosis in CKD. However, the molecular mechanisms leading to mitochondrial fragmentation remain to be elucidated. The present study investigated the role of regulators of calcineurin 1 (RCAN1) in mitochondrial fission and renal interstitial fibrosis using conditional knockout mice in which RCAN1 was genetically deleted in tubular epithelial cells (TECs). TEC-specific deletion of RCAN1 attenuated tubulointerstitial fibrosis and epithelial to mesenchymal transition (EMT)-like phenotype change after unilateral ureteral obstruction (UUO) and ischemia reperfusion injury (IRI) through suppressing TGF-β1/Smad3 signaling pathway. TEC-specific deletion of RCAN1 also reduced the tubular apoptosis after UUO by inhibiting cytochrome c/caspase-9 pathway. Ultrastructure analysis revealed a marked decrease in mitochondrial fragmentation in TECs of RCAN1-deficient mice in experimental CKD models. The expression of mitochondrial profission proteins dynamin-related protein 1 (Drp1) and mitochondrial fission factor (Mff) was also downregulated in obstructed kidney of TEC-specific RCAN1-deficient mice. Furthermore, TEC-specific deletion of RCAN1 attenuated the dysfunctional tubular autophagy by regulating PINK1/Parkin-induced mitophagy in CKD. RCAN1 knockdown and knockout similarly improved the mitochondrial quality control in HK-2 cells and primary cultured mouse tubular cells stimulated by TGF-β1. Put together, our data indicated that RCAN1 plays an important role in the progression of tubulointerstitial fibrosis through regulating the mitochondrial quality. Therefore, targeting RCAN1 may provide a potential therapeutic approach in CKD.
Collapse
Affiliation(s)
- Xue-Yu Sang
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Jing-Jie Xiao
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Qing Liu
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Rui Zhu
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Jia-Jia Dai
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Cheng Zhang
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Hong Yu
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| | - Si-Jun Yang
- ABSL-3 Laboratory at the Center for Animal Experiment, State Key Laboratory of Virology, Wuhan University School of Medicine, Wuhan, P.R. China
| | - Bai-Fang Zhang
- Department of Biochemistry, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China
| |
Collapse
|
9
|
RCAN1 Inhibits BACE2 Turnover by Attenuating Proteasome-Mediated BACE2 Degradation. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1920789. [PMID: 32566665 PMCID: PMC7293731 DOI: 10.1155/2020/1920789] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 05/22/2020] [Indexed: 11/23/2022]
Abstract
Amyloid-β protein (Aβ) is the main component of neuritic plaques, the pathological hallmark of Alzheimer's disease (AD). β-site APP cleaving enzyme 1 (BACE1) is a major β-secretase contributing to Aβ generation. β-site APP cleaving enzyme 2 (BACE2), the homolog of BACE1, is not only a θ-secretase but also a conditional β-secretase. Previous studies showed that regulator of calcineurin 1 (RCAN1) is markedly increased by AD and promotes BACE1 expression. However, the role of RCAN1 in BACE2 regulation remains elusive. Here, we showed that RCAN1 increases BACE2 protein levels. Moreover, RCAN1 inhibits the turnover of BACE2 protein. Furthermore, RCAN1 attenuates proteasome-mediated BACE2 degradation, but not lysosome-mediated BACE2 degradation. Taken together, our work indicates that RCAN1 inhibits BACE2 turnover by attenuating proteasome-mediated BACE2 degradation. It advances our understanding of BACE2 regulation and provides a potential mechanism of BACE2 dysregulation in AD.
Collapse
|
10
|
Pomatto LCD, Davies KJA. The role of declining adaptive homeostasis in ageing. J Physiol 2017; 595:7275-7309. [PMID: 29028112 PMCID: PMC5730851 DOI: 10.1113/jp275072] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 09/01/2017] [Indexed: 12/12/2022] Open
Abstract
Adaptive homeostasis is "the transient expansion or contraction of the homeostatic range for any given physiological parameter in response to exposure to sub-toxic, non-damaging, signalling molecules or events, or the removal or cessation of such molecules or events" (Davies, 2016). Adaptive homeostasis enables biological systems to make continuous short-term adjustments for optimal functioning despite ever-changing internal and external environments. Initiation of adaptation in response to an appropriate signal allows organisms to successfully cope with much greater, normally toxic, stresses. These short-term responses are initiated following effective signals, including hypoxia, cold shock, heat shock, oxidative stress, exercise-induced adaptation, caloric restriction, osmotic stress, mechanical stress, immune response, and even emotional stress. There is now substantial literature detailing a decline in adaptive homeostasis that, unfortunately, appears to manifest with ageing, especially in the last third of the lifespan. In this review, we present the hypothesis that one hallmark of the ageing process is a significant decline in adaptive homeostasis capacity. We discuss the mechanistic importance of diminished capacity for short-term (reversible) adaptive responses (both biochemical and signal transduction/gene expression-based) to changing internal and external conditions, for short-term survival and for lifespan and healthspan. Studies of cultured mammalian cells, worms, flies, rodents, simians, apes, and even humans, all indicate declining adaptive homeostasis as a potential contributor to age-dependent senescence, increased risk of disease, and even mortality. Emerging work points to Nrf2-Keap1 signal transduction pathway inhibitors, including Bach1 and c-Myc, both of whose tissue concentrations increase with age, as possible major causes for age-dependent loss of adaptive homeostasis.
Collapse
Affiliation(s)
- Laura C. D. Pomatto
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology CenterUniversity of Southern CaliforniaLos AngelesCA 90089USA
| | - Kelvin J. A. Davies
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology CenterUniversity of Southern CaliforniaLos AngelesCA 90089USA
- Molecular and Computational Biology Program, Department of Biological Sciences of the Dornsife College of LettersArts & Sciences: the University of Southern CaliforniaLos AngelesCA 90089‐0191USA
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Regulator of Calcineurin 3 Ameliorates Autoimmune Arthritis by Suppressing Th17 Cell Differentiation. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2034-2045. [PMID: 28704638 DOI: 10.1016/j.ajpath.2017.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/10/2017] [Accepted: 05/15/2017] [Indexed: 01/10/2023]
Abstract
Regulator of calcineurin 3 (RCAN3), an endogenous regulator of the calcineurin-nuclear factor of activated T cells (NFAT) signaling pathway, inhibits the phosphatase activity of calcineurin, the nuclear translocation of NFAT, and the NFAT downstream pathway. To investigate the effects of RCAN3 on T-cell regulatory function and the development and progression of inflammatory arthritis, we studied the effects of RCAN3 transfection on regulation of Th17 cell differentiation in a murine T-lymphoma cell line and primary splenic CD4+ T cells. Overexpression of RCAN3 suppressed Th17 cell differentiation through the down-regulation of RAR receptor orphan receptor γT mRNA and up-regulation of forkhead box P3 mRNA. In mice with collagen-induced arthritis, injection of an RCAN3-overexpression vector controlled arthritis development in vivo. Injection of RCAN3 reduced the formation of osteoclasts and expression of inflammatory cytokines in vivo. Antioxidants stimulated the expression of RCAN3 in vitro, and combination therapy with pcDNA-RCAN3 had a synergistic suppressive effect on the development of arthritis. These data suggest that RCAN3 may be an effective treatment for rheumatoid arthritis.
Collapse
|
14
|
Asai M, Kinjo A, Kimura S, Mori R, Kawakubo T, Shirotani K, Yagishita S, Maruyama K, Iwata N. Perturbed Calcineurin-NFAT Signaling Is Associated with the Development of Alzheimer's Disease. Biol Pharm Bull 2017; 39:1646-1652. [PMID: 27725441 DOI: 10.1248/bpb.b16-00350] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Down syndrome (DS), the most common genetic disorder, is caused by trisomy 21. DS is accompanied by heart defects, hearing and vision problems, obesity, leukemia, and other conditions, including Alzheimer's disease (AD). In comparison, most cancers are rare in people with DS. Overexpression of dual specificity tyrosine-phosphorylation-regulated kinase 1A and a regulator of calcineurin 1 located on chromosome 21 leads to excessive suppression of the calcineurin-nuclear factor of activated T cells (NFAT) signaling pathway, resulting in reduced expression of a critical angiogenic factor. However, it is unclear whether the calcineurin-NFAT signaling pathway is involved in AD pathology in DS patients. Here, we investigated the association between the calcineurin-NFAT signaling pathway and AD using neuronal cells. Short-term pharmacological stimulation decreased gene expression of tau and neprilysin, and long-term inhibition of the signaling pathway decreased that of amyloid precursor protein. Moreover, a calcineurin inhibitor, cyclosporine A, also decreased neprilysin activity, leading to increases in amyloid-β peptide levels. Taken together, our results suggest that a dysregulation in calcineurin-NFAT signaling may contribute to the early onset of AD in people with DS.
Collapse
Affiliation(s)
- Masashi Asai
- School of Pharmaceutical Sciences, Nagasaki University
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Bonet-Costa V, Pomatto LCD, Davies KJA. The Proteasome and Oxidative Stress in Alzheimer's Disease. Antioxid Redox Signal 2016; 25:886-901. [PMID: 27392670 PMCID: PMC5124752 DOI: 10.1089/ars.2016.6802] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SIGNIFICANCE Alzheimer's disease is a neurodegenerative disorder that is projected to exceed more than 100 million cases worldwide by 2050. Aging is considered the primary risk factor for some 90% of Alzheimer's cases but a significant 10% of patients suffer from aggressive, early-onset forms of the disease. There is currently no effective Alzheimer's treatment and this, coupled with a growing aging population, highlights the necessity to understand the mechanism(s) of disease initiation and propagation. A major hallmark of Alzheimer's disease pathology is the accumulation of amyloid-β (Aβ) aggregates (an early marker of Alzheimer's disease), and neurofibrillary tangles, comprising the hyper-phosphorylated microtubule-associated protein Tau. Recent Advances: Protein oxidation is frequently invoked as a potential factor in the progression of Alzheimer's disease; however, whether it is a cause or a consequence of the pathology is still being debated. The Proteasome complex is a major regulator of intracellular protein quality control and an essential proteolytic enzyme for the processing of both Aβ and Tau. Recent studies have indicated that both protein oxidation and excessive phosphorylation may limit Proteasomal processing of Aβ and Tau in Alzheimer's disease. CRITICAL ISSUES Thus, the Proteasome may be a key factor in understanding the development of Alzheimer's disease pathology; however, its significance is still very much under investigation. FUTURE DIRECTIONS Discovering how the proteasome is affected, regulated, or dysregulated in Alzheimer's disease could be a valuable tool in the efforts to understand and, ultimately, eradicate the disease. Antioxid. Redox Signal. 25, 886-901.
Collapse
Affiliation(s)
- Vicent Bonet-Costa
- Leonard Davis School of Gerontology, Ethel Percy Andrus Gerontology Center, The Division of Molecular and Computational Biology, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, The University of Southern California , Los Angeles, California
| | - Laura Corrales-Diaz Pomatto
- Leonard Davis School of Gerontology, Ethel Percy Andrus Gerontology Center, The Division of Molecular and Computational Biology, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, The University of Southern California , Los Angeles, California
| | - Kelvin J A Davies
- Leonard Davis School of Gerontology, Ethel Percy Andrus Gerontology Center, The Division of Molecular and Computational Biology, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, The University of Southern California , Los Angeles, California
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Emrani R, Rébillard A, Lefeuvre L, Gratas-Delamarche A, Davies KJA, Cillard J. The calcineurin antagonist RCAN1-4 is induced by exhaustive exercise in rat skeletal muscle. Free Radic Biol Med 2015; 87:290-9. [PMID: 26122706 DOI: 10.1016/j.freeradbiomed.2015.06.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/18/2015] [Accepted: 06/21/2015] [Indexed: 12/12/2022]
Abstract
The aim of this work was to study the regulation of the calcineurin antagonist regulator of calcineurin 1 (RCAN1) in rat skeletal muscles after exhaustive physical exercise, which is a physiological modulator of oxidative stress. Three skeletal muscles, namely extensor digitorum longus (EDL), gastrocnemius, and soleus, were investigated. Exhaustive exercise increased RCAN1-4 protein levels in EDL and gastrocnemius, but not in soleus. Protein oxidation as an index of oxidative stress was increased in EDL and gastrocnemius, but remained unchanged in soleus. However, lipid peroxidation was increased in all three muscles. CuZnSOD and catalase protein levels were increased at 3 h postexercise in soleus, whereas they remained unchanged in EDL and gastrocnemius. Calcineurin enzymatic activity declined in EDL and gastrocnemius but not in soleus, and its protein expression was decreased in all three muscles. The level of PGC1-α protein remained unchanged, whereas the protein expression of the transcription factor NFATc4 was decreased in all three muscles. Adiponectin expression was increased in all three muscles. RCAN1-4 expression in EDL and gastrocnemius muscles was augmented by the oxidative stress generated from exhaustive exercise. We propose that increased RCAN1-4 expression and the signal transduction pathways it regulates represent important components of the physiological adaptation to exercise-induced oxidative stress.
Collapse
Affiliation(s)
- Ramin Emrani
- Laboratory of Movement, Sport & Health Sciences (EA 1274), Faculty of Pharmacy, University Rennes 1, 35043 Rennes Cédex, France
| | - Amélie Rébillard
- Laboratory of Movement, Sport & Health Sciences, University Rennes 2, Ecole Normale Supérieure Rennes, 35170 Bruz, France
| | - Luz Lefeuvre
- Laboratory of Movement, Sport & Health Sciences, University Rennes 2, Ecole Normale Supérieure Rennes, 35170 Bruz, France
| | - Arlette Gratas-Delamarche
- Laboratory of Movement, Sport & Health Sciences, University Rennes 2, Ecole Normale Supérieure Rennes, 35170 Bruz, France
| | - Kelvin J A Davies
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, and Division of Molecular and Computational Biology, Department of Biological Sciences, Dornsife College of Letters, Arts & Sciences, University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Josiane Cillard
- Laboratory of Movement, Sport & Health Sciences (EA 1274), Faculty of Pharmacy, University Rennes 1, 35043 Rennes Cédex, France.
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
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
|
22
|
Bradford D, Tschanz JT, Smith KR, Østbye T, Corcoran C, Welsh-Bohmer KA, Norton MC, For the Cache County Investigators. Impact of offspring death on cognitive health in late life: the Cache County study. Am J Geriatr Psychiatry 2014; 22:1307-15. [PMID: 23954042 PMCID: PMC3923854 DOI: 10.1016/j.jagp.2013.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 04/21/2013] [Accepted: 05/09/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Experiencing the death of a child is associated with negative short-term mental health consequences, but less is known about cognitive outcomes and whether such associations extend to late life. We tested the hypothesis that experiencing an offspring death (OD) is associated with an increased rate of cognitive decline in late life. METHODS This population-based longitudinal study observed four cognitive statuses spaced 3-4 years apart, linked to an extensive database containing objective genealogic and vital statistics data. Home visits were conducted with 3,174 residents of a rural county in northern Utah, initially without dementia, aged 65-105. Cognitive status was measured with the Modified Mini-Mental State Exam at baseline and at 3-, 7-, and 10-year follow-ups. OD was obtained from the Utah Population Database, which contains statewide birth and death records. RESULTS In linear mixed models, controlling for age, gender, education, and apolipoprotein E status, subjects who experienced OD while younger than age 31 years experienced a significantly faster rate of cognitive decline in late life, but only if they had an ε4 allele. Reclassifying all OD (regardless of age) according to subsequent birth of another child, OD was only related to faster cognitive decline when there were no subsequent births. CONCLUSION Experiencing OD in early adulthood has a long-term association with cognitive functioning in late life, with a gene-environment interaction at the apolipoprotein E locus. Subsequent birth of another child attenuates this association.
Collapse
Affiliation(s)
- Daylee Bradford
- Department of Family Consumer and Human Development, Utah State University
| | - JoAnn T. Tschanz
- Department of Psychology, Center for Epidemiologic Studies, Utah State University
| | - Ken R. Smith
- Department of Family and Consumer Studies; Population Sciences, Huntsman Cancer Institute, University of Utah
| | - Truls Østbye
- Duke Global Health Institute, Duke University, and Duke – NUS Graduate Medical School, Singapore
| | - Chris Corcoran
- Department of Mathematics and Statistics, Center for Epidemiologic Studies, Utah State University
| | | | - Maria C. Norton
- Department of Family Consumer and Human Development, Utah State University,Department of Psychology, Center for Epidemiologic Studies, Utah State University
| | | |
Collapse
|
23
|
Montoya JC, Fajardo D, Peña A, Sánchez A, Domínguez MC, Satizábal JM, García-Vallejo F. Global differential expression of genes located in the Down Syndrome Critical Region in normal human brain. Colomb Med (Cali) 2014; 45:154-61. [PMID: 25767303 PMCID: PMC4350380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 11/10/2014] [Accepted: 12/19/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The information of gene expression obtained from databases, have made possible the extraction and analysis of data related with several molecular processes involving not only in brain homeostasis but its disruption in some neuropathologies; principally in Down syndrome and the Alzheimer disease. OBJECTIVE To correlate the levels of transcription of 19 genes located in the Down Syndrome Critical Region (DSCR) with their expression in several substructures of normal human brain. METHODS There were obtained expression profiles of 19 DSCR genes in 42 brain substructures, from gene expression values available at the database of the human brain of the Brain Atlas of the Allen Institute for Brain Sciences", (http://human.brain-map.org/). The co-expression patterns of DSCR genes in brain were calculated by using multivariate statistical methods. RESULTS Highest levels of gene expression were registered at caudate nucleus, nucleus accumbens and putamen among central areas of cerebral cortex. Increased expression levels of RCAN1 that encode by a protein involved in signal transduction process of the CNS were recorded for PCP4 that participates in the binding to calmodulin and TTC3; a protein that is associated with differentiation of neurons. That previously identified brain structures play a crucial role in the learning process, in different class of memory and in motor skills. CONCLUSION The precise regulation of DSCR gene expression is crucial to maintain the brain homeostasis, especially in those areas with high levels of gene expression associated with a remarkable process of learning and cognition.
Collapse
Affiliation(s)
- Julio Cesar Montoya
- Department of Physiological Sciences, School of Basic Sciences, Faculty of Health, Universidad del Valle. Cali, Colombia, Faculty of Basic Sciences, Universidad Autónoma de Occidente, Cali, Colombia
| | - Dianora Fajardo
- Laboratory of Molecular Biology and Pathogenesis LABIOMOL. Universidad del Valle, Cali, Colombia
| | - Angela Peña
- Laboratory of Molecular Biology and Pathogenesis LABIOMOL. Universidad del Valle, Cali, Colombia
| | - Adalberto Sánchez
- Department of Physiological Sciences, School of Basic Sciences, Faculty of Health, Universidad del Valle. Cali, Colombia
| | - Martha C Domínguez
- Department of Physiological Sciences, School of Basic Sciences, Faculty of Health, Universidad del Valle. Cali, Colombia, Laboratory of Molecular Biology and Pathogenesis LABIOMOL. Universidad del Valle, Cali, Colombia
| | - José María Satizábal
- Department of Physiological Sciences, School of Basic Sciences, Faculty of Health, Universidad del Valle. Cali, Colombia
| | - Felipe García-Vallejo
- Department of Physiological Sciences, School of Basic Sciences, Faculty of Health, Universidad del Valle. Cali, Colombia, Laboratory of Molecular Biology and Pathogenesis LABIOMOL. Universidad del Valle, Cali, Colombia
| |
Collapse
|
24
|
Malakooti N, Pritchard MA, Adlard PA, Finkelstein DI. Role of metal ions in the cognitive decline of Down syndrome. Front Aging Neurosci 2014; 6:136. [PMID: 25002847 PMCID: PMC4066992 DOI: 10.3389/fnagi.2014.00136] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 06/09/2014] [Indexed: 11/17/2022] Open
Abstract
Down syndrome (DS), caused by trisomy of whole or part of chromosome 21 is the most common mental impairment. All people with DS suffer from cognitive decline and develop Alzheimer’s disease (AD) by the age of 40. The appearance of enlarged early endosomes, followed by Amyloid βpeptide deposition, the appearance of tau-containing neurofibrillary tangles and basal forebrain cholinergic neuron (BFCN) degeneration are the neuropathological characteristics of this disease. In this review we will examine the role of metal ion dyshomeostasis and the genes which may be involved in these processes, and relate these back to the manifestation of age-dependent cognitive decline in DS.
Collapse
Affiliation(s)
- Nakisa Malakooti
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health Parkville, VIC, Australia
| | | | - Paul A Adlard
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health Parkville, VIC, Australia
| | - David I Finkelstein
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health Parkville, VIC, Australia
| |
Collapse
|
25
|
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
|
26
|
Wu Y, Ly PTT, Song W. Aberrant expression of RCAN1 in Alzheimer's pathogenesis: a new molecular mechanism and a novel drug target. Mol Neurobiol 2014; 50:1085-97. [PMID: 24752590 DOI: 10.1007/s12035-014-8704-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 03/31/2014] [Indexed: 01/08/2023]
Abstract
AD, a devastating neurodegenerative disorder, is the most common cause of dementia in the elderly. Patients with AD are characterized by three hallmarks of neuropathology including neuritic plaque deposition, neurofibrillary tangle formation, and neuronal loss. Growing evidences indicate that dysregulation of regulator of calcineurin 1 (RCAN1) plays an important role in the pathogenesis of AD. Aberrant RCAN1 expression facilitates neuronal apoptosis and Tau hyperphosphorylation, leading to neuronal loss and neurofibrillary tangle formation. This review aims to describe the recent advances of the regulation of RCAN1 expression and its physiological functions. Moreover, the AD risk factors-induced RCAN1 dysregulation and its role in promoting neuronal loss, synaptic impairments and neurofibrillary tangle formation are summarized. Furthermore, we provide an outlook into the effects of RCAN1 dysregulation on APP processing, Aβ generation and neuritic plaque formation, and the possible underlying mechanisms, as well as the potential of targeting RCAN1 as a new therapeutic approach.
Collapse
Affiliation(s)
- Yili Wu
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, Graduate Program in Neuroscience, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, Canada, V6T 1Z3
| | | | | |
Collapse
|
27
|
Sun X, Wu Y, Herculano B, Song W. RCAN1 overexpression exacerbates calcium overloading-induced neuronal apoptosis. PLoS One 2014; 9:e95471. [PMID: 24751678 PMCID: PMC3994074 DOI: 10.1371/journal.pone.0095471] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 03/26/2014] [Indexed: 01/23/2023] Open
Abstract
Down Syndrome (DS) patients develop characteristic Alzheimer's Disease (AD) neuropathology after their middle age. Prominent neuronal loss has been observed in the cortical regions of AD brains. However, the underlying mechanism leading to this neuronal loss in both DS and AD remains to be elucidated. Calcium overloading and oxidative stress have been implicated in AD pathogenesis. Two major isoforms of regulator of calcineurin 1 (RCAN1), RCAN1.1 and RCAN1.4, are detected in human brains. In this report we defined the transcriptional regulation of RCAN1.1 and RCAN1.4 by two alternative promoters. Calcium overloading upregulated RCAN1.4 expression by activating RCAN1.4 promoter through calcineurin-NFAT signaling pathway, thus forming a negative feedback loop in isoform 4 regulation. Furthermore, RCAN1.4 overexpression exacerbated calcium overloading-induced neuronal apoptosis, which was mediated by caspase-3 apoptotic pathway. Our results suggest that downregulating RCAN1.4 expression in neurons could be beneficial to AD patients.
Collapse
Affiliation(s)
- Xiulian Sun
- Qilu Hospital of Shandong University, Jinan, China
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada
| | - Yili Wu
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, and Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada
| | - Bruno Herculano
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada
| | - Weihong Song
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, and Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada
- * E-mail:
| |
Collapse
|
28
|
Perluigi M, Pupo G, Tramutola A, Cini C, Coccia R, Barone E, Head E, Butterfield DA, Di Domenico F. Neuropathological role of PI3K/Akt/mTOR axis in Down syndrome brain. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1144-53. [PMID: 24735980 DOI: 10.1016/j.bbadis.2014.04.007] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/19/2014] [Accepted: 04/07/2014] [Indexed: 01/11/2023]
Abstract
Down syndrome (DS) is the most frequent genetic cause of intellectual disability characterized by the presence of three copies of chromosome 21 (Chr21). Individuals with DS have sufficient neuropathology for a diagnosis of Alzheimer's disease (AD) after the age of 40years. The aim of our study is to gain new insights in the molecular mechanisms impaired in DS subjects that eventually lead to the development of dementia. We evaluate the PI3K/Akt/mTOR axis in the frontal cortex from DS cases (under the age of 40years) and DS with AD neuropathology compared with age-matched controls (Young and Old). The PI3K/Akt/mTOR axis may control several key pathways involved in AD that, if aberrantly regulated, affect amyloid beta (Aβ) deposition and tau phosphorylation. Our results show a hyperactivation of PI3K/Akt/mTOR axis in individuals with DS, with and without AD pathology, in comparison with respective controls. The PI3K/Akt/mTOR deregulation results in decreased autophagy, inhibition of IRS1 and GSK3β activity. Moreover, our data suggest that aberrant activation of the PI3K/Akt/mTOR axis acts in parallel to RCAN1 in phosphorylating tau, in DS and DS/AD. In conclusion, this study provides insights into the neuropathological mechanisms that may be engaged during the development of AD in DS. We suggest that deregulation of this signaling cascade is already evident in young DS cases and persist in the presence of AD pathology. The impairment of the PI3K/Akt/mTOR axis in DS population might represent a key-contributing factor to the neurodegenerative process that culminates in Alzheimer-like dementia.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Gilda Pupo
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Chiara Cini
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Raffaella Coccia
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Elizabeth Head
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA
| | - D Allan Butterfield
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA; Department of Chemistry and Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506-0055, USA
| | - Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| |
Collapse
|
29
|
Serrano-Candelas E, Farré D, Aranguren-Ibáñez Á, Martínez-Høyer S, Pérez-Riba M. The vertebrate RCAN gene family: novel insights into evolution, structure and regulation. PLoS One 2014; 9:e85539. [PMID: 24465593 PMCID: PMC3896409 DOI: 10.1371/journal.pone.0085539] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 12/04/2013] [Indexed: 12/30/2022] Open
Abstract
Recently there has been much interest in the Regulators of Calcineurin (RCAN) proteins which are important endogenous modulators of the calcineurin-NFATc signalling pathway. They have been shown to have a crucial role in cellular programmes such as the immune response, muscle fibre remodelling and memory, but also in pathological processes such as cardiac hypertrophy and neurodegenerative diseases. In vertebrates, the RCAN family form a functional subfamily of three members RCAN1, RCAN2 and RCAN3 whereas only one RCAN is present in the rest of Eukarya. In addition, RCAN genes have been shown to collocate with RUNX and CLIC genes in ACD clusters (ACD21, ACD6 and ACD1). How the RCAN genes and their clustering in ACDs evolved is still unknown. After analysing RCAN gene family evolution using bioinformatic tools, we propose that the three RCAN vertebrate genes within the ACD clusters, which evolved from single copy genes present in invertebrates and lower eukaryotes, are the result of two rounds of whole genome duplication, followed by a segmental duplication. This evolutionary scenario involves the loss or gain of some RCAN genes during evolution. In addition, we have analysed RCAN gene structure and identified the existence of several characteristic features that can be involved in RCAN evolution and gene expression regulation. These included: several transposable elements, CpG islands in the 5′ region of the genes, the existence of antisense transcripts (NAT) associated with the three human genes, and considerable evidence for bidirectional promoters that regulate RCAN gene expression. Furthermore, we show that the CpG island associated with the RCAN3 gene promoter is unmethylated and transcriptionally active. All these results provide timely new insights into the molecular mechanisms underlying RCAN function and a more in depth knowledge of this gene family whose members are obvious candidates for the development of future therapies.
Collapse
Affiliation(s)
- Eva Serrano-Candelas
- Cancer and Human Molecular Genetics Department, Bellvitge Biomedical Research Institute – IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Domènec Farré
- Biological Aggression and Response Mechanisms Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer – IDIBAPS, Barcelona, Spain
| | - Álvaro Aranguren-Ibáñez
- Cancer and Human Molecular Genetics Department, Bellvitge Biomedical Research Institute – IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Sergio Martínez-Høyer
- Cancer and Human Molecular Genetics Department, Bellvitge Biomedical Research Institute – IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Mercè Pérez-Riba
- Cancer and Human Molecular Genetics Department, Bellvitge Biomedical Research Institute – IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
- * E-mail:
| |
Collapse
|
30
|
Ermak G, Davies KJA. Chronic high levels of the RCAN1-1 protein may promote neurodegeneration and Alzheimer disease. Free Radic Biol Med 2013; 62:47-51. [PMID: 23369757 PMCID: PMC4720382 DOI: 10.1016/j.freeradbiomed.2013.01.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 12/28/2012] [Accepted: 01/17/2013] [Indexed: 02/07/2023]
Abstract
The RCAN1 gene encodes three different protein isoforms: RCAN1-4, RCAN1-1L, and RCAN1-1S. RCAN1-1L is the RCAN1 isoform predominantly expressed in human brains. RCAN1 proteins have been shown to regulate various other proteins and cellular functions, including calcineurin, glycogen synthase kinase-3β (GSK-3β), the mitochondrial adenine nucleotide transporter (ANT), stress adaptation, ADP/ATP exchange in mitochondria, and the mitochondrial permeability transition pore (mtPTP). The effects of increased RCAN1 gene expression seem to depend both on the specific RCAN1 protein isoform(s) synthesized and on the length of time the level of each isoform is elevated. Transiently elevated RCAN1-4 and RCAN1-1L protein levels, lasting just a few hours, can be neuroprotective under acute stress conditions, including acute oxidative stress. We propose that, by transiently inhibiting the phosphatase calcineurin, RCAN1-4 and RCAN1-1L may reinforce and extend protective stress-adaptive cell responses. In contrast, prolonged elevation of RCAN1-1L levels is associated with the types of neurodegeneration observed in several diseases, including Alzheimer disease and Down syndrome. RCAN1-1L levels can also be increased by multiple chronic stresses and by glucocorticoids, both of which can cause neurodegeneration. Although increasing levels of RCAN1-1L for just a few months has no overtly obvious neurodegenerative effect, it does suppress neurogenesis. Longer term elevation of RCAN1-1L levels (for at least 16 months), however, can lead to the first signs of neurodegeneration. Such neurodegeneration may be precipitated by (RCAN1-1L-mediated) prolonged calcineurin inhibition and GSK-3β induction/activation, both of which promote tau hyperphosphorylation, and/or by (RCAN1-1L-mediated) effects on the mitochondrial ANT, diminished ATP/ADP ratio, opening of the mtPTP, and mitochondrial autophagy. We propose that RCAN1-1L operates through various molecular mechanisms, primarily dependent upon the length of time protein levels are elevated. We also suggest that models analyzing long-term RCAN1 gene overexpression may help us to understand the molecular mechanisms of neurodegeneration in diseases such as Alzheimer disease, Down syndrome, and possibly others.
Collapse
Affiliation(s)
- Gennady Ermak
- Ethel Percy Andrus Gerontology Center, Davis School of Gerontology, and Division of Molecular & Computational Biology, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Kelvin J A Davies
- Ethel Percy Andrus Gerontology Center, Davis School of Gerontology, and Division of Molecular & Computational Biology, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA 90089-0191, USA.
| |
Collapse
|
31
|
Bushman DM, Chun J. The genomically mosaic brain: aneuploidy and more in neural diversity and disease. Semin Cell Dev Biol 2013; 24:357-369. [PMID: 23466288 PMCID: PMC3637860 DOI: 10.1016/j.semcdb.2013.02.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 01/25/2013] [Accepted: 02/06/2013] [Indexed: 10/27/2022]
Abstract
Genomically identical cells have long been assumed to comprise the human brain, with post-genomic mechanisms giving rise to its enormous diversity, complexity, and disease susceptibility. However, the identification of neural cells containing somatically generated mosaic aneuploidy - loss and/or gain of chromosomes from a euploid complement - and other genomic variations including LINE1 retrotransposons and regional patterns of DNA content variation (DCV), demonstrate that the brain is genomically heterogeneous. The precise phenotypes and functions produced by genomic mosaicism are not well understood, although the effects of constitutive aberrations, as observed in Down syndrome, implicate roles for defined mosaic genomes relevant to cellular survival, differentiation potential, stem cell biology, and brain organization. Here we discuss genomic mosaicism as a feature of the normal brain as well as a possible factor in the weak or complex genetic linkages observed for many of the most common forms of neurological and psychiatric diseases.
Collapse
Affiliation(s)
- Diane M. Bushman
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, USA
- Biomedical Sciences Graduate Program, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Jerold Chun
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
32
|
Saavedra-Rodríguez L, Feig LA. Chronic social instability induces anxiety and defective social interactions across generations. Biol Psychiatry 2013; 73:44-53. [PMID: 22906514 PMCID: PMC3826464 DOI: 10.1016/j.biopsych.2012.06.035] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 06/05/2012] [Accepted: 06/23/2012] [Indexed: 01/06/2023]
Abstract
BACKGROUND Chronic social instability during adolescence and early adulthood is known to produce a variety of long-lasting effects that may contribute to future psychiatric disorders. However, its potential to affect future generations has not been tested. METHODS Female and male mice were exposed to chronic social stress involving social instability and disruption of social hierarchy from postnatal day 27 to 76. After treatment, a group of animals was used to evaluate long-term behavioral effects of the stress exposure, and other mice were used to generate F1, F2, and F3 offspring, to test for behavioral effects across generations. RESULTS Chronic social instability during adolescence and early adulthood induces persistent behavioral alterations, including enhanced anxiety and social deficits that are transmitted predominantly to females across at least three generations. Both mothers and fathers can transmit all of these altered behaviors to their F1 offspring. However, only F1 fathers transmit all of them to their F2 and F3 daughters. In the F1 generation, enhanced anxiety and social deficits are associated with elevated serum corticosterone levels; however, in the F2 and F3 generations, they are not. CONCLUSIONS These findings support the idea that individual risk for psychiatric disorders that involve enhanced anxiety and/or social dysfunction may be dependent not only on the specific alleles of genes that are inherited from one's parents and on one's own experiences, but also on the experiences of one's parents when they were young.
Collapse
|
33
|
Wu Y, Song W. Regulation of RCAN1 translation and its role in oxidative stress-induced apoptosis. FASEB J 2012; 27:208-21. [PMID: 23038757 DOI: 10.1096/fj.12-213124] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Abnormal expression of regulator of calcineurin 1 (RCAN1) has been implicated in Alzheimer's disease (AD) and Down's syndrome (DS). There are two major isoforms of RCAN1, isoforms 1 and 4. RCAN1 isoform 1 is predominantly expressed in the brain, particularly in neurons. In this report, we showed that there are two translation start codons in RCAN1 exon 1 serving as a functional translation initiation site to generate a longer 41-kDa isoform 1 (RCAN1.1L) and a shorter 31-kDa isoform 1 (RCAN1.1S). The first translation initiation site has higher translation efficiency than the downstream second one, and the translation initiation of two AUG sites is by a Cap-dependent mechanism. Short-term expression of RCAN1.1L protected SH-SY5Y cells from oxidative stress-induced apoptosis by inhibiting caspase-3 activation. However, long-term accumulation of RCAN1.1L in SH-SY5Y cells promoted oxidative stress-induced apoptosis via caspase-3 activation, and terminal deoxynucleotidyl transferase dUTP nick end labeling assay showed that the apoptosis ratio was increased to 499.03 ± 47.56% in SH-1.1L cells compared with 283.93 ± 28.66% in control cells. Furthermore, we found that RCAN1.1L is significantly elevated in the AD brains and patients with DS. RCAN1.1S is expressed at a low level in both human cells and brain tissues. Our results defined the regulatory mechanism underlying RCAN1 expression and the roles of RCAN1.1 in oxidative stress-induced neurodegeneration in AD and DS pathogenesis.
Collapse
Affiliation(s)
- Yili Wu
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | | |
Collapse
|
34
|
Molecular and cellular alterations in Down syndrome: toward the identification of targets for therapeutics. Neural Plast 2012; 2012:171639. [PMID: 22848846 PMCID: PMC3403492 DOI: 10.1155/2012/171639] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 04/18/2012] [Accepted: 04/19/2012] [Indexed: 12/25/2022] Open
Abstract
Down syndrome is a complex disease that has challenged molecular and cellular research for more than 50 years. Understanding the molecular bases of morphological, cellular, and functional alterations resulting from the presence of an additional complete chromosome 21 would aid in targeting specific genes and pathways for rescuing some phenotypes. Recently, progress has been made by characterization of brain alterations in mouse models of Down syndrome. This review will highlight the main molecular and cellular findings recently described for these models, particularly with respect to their relationship to Down syndrome phenotypes.
Collapse
|
35
|
Ermak G, Sojitra S, Yin F, Cadenas E, Cuervo AM, Davies KJA. Chronic expression of RCAN1-1L protein induces mitochondrial autophagy and metabolic shift from oxidative phosphorylation to glycolysis in neuronal cells. J Biol Chem 2012; 287:14088-98. [PMID: 22389495 DOI: 10.1074/jbc.m111.305342] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Expression of the RCAN1 gene can be induced by multiple stresses. RCAN1 proteins (RCAN1s) have both protective and harmful effects and are implicated in common human pathologies. The mechanisms by which RCAN1s function, however, remain poorly understood. We identify RCAN1s as regulators of mitochondrial autophagy (mitophagy) and demonstrate that induction of RCAN1-1L can cause dramatic degradation of mitochondria. The mechanisms of such degradation involve the adenine nucleotide translocator and mitochondrial permeability transition pore opening. We also demonstrate that RCAN1-1L induction can shift cellular bioenergetics from aerobic respiration to glycolysis, yet RCAN1-1L has very little effect on cell division, whereas it has a cumulative negative effect on cell survival. These results shed the light on mechanisms by which RCAN1s can protect or harm cells and by which they may operate in human pathologies. They also suggest that RCAN1s are important players in autophagy and such elusive phenomena as the mitochondrial permeability transition pore.
Collapse
Affiliation(s)
- Gennady Ermak
- Ethel Percy Andrus Gerontology Center of the Davis School of Gerontology and the Division of Molecular and Computational Biology, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California 90089-0191, USA
| | | | | | | | | | | |
Collapse
|