51
|
Moos WH, Faller DV, Glavas IP, Harpp DN, Kanara I, Mavrakis AN, Pernokas J, Pernokas M, Pinkert CA, Powers WR, Sampani K, Steliou K, Vavvas DG, Zamboni RJ, Kodukula K, Chen X. Klotho Pathways, Myelination Disorders, Neurodegenerative Diseases, and Epigenetic Drugs. Biores Open Access 2020; 9:94-105. [PMID: 32257625 PMCID: PMC7133426 DOI: 10.1089/biores.2020.0004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In this review we outline a rationale for identifying neuroprotectants aimed at inducing endogenous Klotho activity and expression, which is epigenetic action, by definition. Such an approach should promote remyelination and/or stimulate myelin repair by acting on mitochondrial function, thereby heralding a life-saving path forward for patients suffering from neuroinflammatory diseases. Disorders of myelin in the nervous system damage the transmission of signals, resulting in loss of vision, motion, sensation, and other functions depending on the affected nerves, currently with no effective treatment. Klotho genes and their single-pass transmembrane Klotho proteins are powerful governors of the threads of life and death, true to the origin of their name, Fates, in Greek mythology. Among its many important functions, Klotho is an obligatory co-receptor that binds, activates, and/or potentiates critical fibroblast growth factor activity. Since the discovery of Klotho a little over two decades ago, it has become ever more apparent that when Klotho pathways go awry, oxidative stress and mitochondrial dysfunction take over, and age-related chronic disorders are likely to follow. The physiological consequences can be wide ranging, potentially wreaking havoc on the brain, eye, kidney, muscle, and more. Central nervous system disorders, neurodegenerative in nature, and especially those affecting the myelin sheath, represent worthy targets for advancing therapies that act upon Klotho pathways. Current drugs for these diseases, even therapeutics that are disease modifying rather than treating only the symptoms, leave much room for improvement. It is thus no wonder that this topic has caught the attention of biomedical researchers around the world.
Collapse
Affiliation(s)
- Walter H. Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, San Francisco, California
- ShangPharma Innovation, Inc., South San Francisco, California
| | - Douglas V. Faller
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Cancer Research Center, Boston University School of Medicine, Boston, Massachusetts
| | - Ioannis P. Glavas
- Department of Ophthalmology, New York University School of Medicine, New York, New York
| | - David N. Harpp
- Department of Chemistry, McGill University, Montreal, Canada
| | | | - Anastasios N. Mavrakis
- Department of Medicine, Tufts University School of Medicine, St. Elizabeth's Medical Center, Boston, Massachusetts
| | - Julie Pernokas
- Advanced Dental Associates of New England, Woburn, Massachusetts
| | - Mark Pernokas
- Advanced Dental Associates of New England, Woburn, Massachusetts
| | - Carl A. Pinkert
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Whitney R. Powers
- Department of Health Sciences, Boston University, Boston, Massachusetts
- Department of Anatomy, Boston University School of Medicine, Boston, Massachusetts
| | - Konstantina Sampani
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Beetham Eye Institute, Joslin Diabetes Center, Boston, Massachusetts
| | - Kosta Steliou
- Cancer Research Center, Boston University School of Medicine, Boston, Massachusetts
- PhenoMatriX, Inc., Natick, Massachusetts
| | - Demetrios G. Vavvas
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | | | | | - Xiaohong Chen
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| |
Collapse
|
52
|
Identification of the cleavage sites leading to the shed forms of human and mouse anti-aging and cognition-enhancing protein Klotho. PLoS One 2020; 15:e0226382. [PMID: 31929539 PMCID: PMC6957300 DOI: 10.1371/journal.pone.0226382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/25/2019] [Indexed: 12/02/2022] Open
Abstract
Klotho is an age-extending, cognition-enhancing protein found to be down-regulated in aged mammals when age-related diseases start to appear. Low levels of Klotho occur in neurodegenerative diseases, kidney disease and many cancers. Many normal and pathologic processes involve the proteolytic shedding of membrane proteins. Transmembrane (TM) Klotho contains two homologous domains, KL1 and KL2 with homology to glycosidases. After shedding by ADAM 10 and 17, a shed Klotho isoform is released into serum and urine by the kidney, and into the CSF by the choroid plexus. We previously reported that human Klotho contains two major cleavage sites. However, the exact cleavage site responsible for the cleavage between the KL1 and KL2 domains remains unknown for the human Klotho, and both sites are unknown for mouse Klotho. In this study, we aimed to identify the cleavage sites leading to the shed forms of human and mouse Klotho. Mutations in the region close to the TM domain of mouse Klotho result in the reduced shedding of the 130 kD (KL1+KL2) and 70 kD (KL1) fragments, suggesting that the cleavage site lies within the mutated region. We further identified the cleavage sites responsible for the cleavage between KL1 and KL2 of human and mouse Klotho. Moreover, mutated Klotho proteins have similar subcellular localization patterns as wild type Klotho. Finally, in an FGF23 functional assay, all Klotho mutants with a nine amino acid deletion can also function as an FGFR1 co-receptor for FGF23 signaling, however, the signaling activity was greatly reduced. The study provides new and important information on Klotho shedding, and paves the way for studies aimed to distinguish between the distinct roles of the various isoforms of Klotho.
Collapse
|
53
|
The goddess who spins the thread of life: Klotho, psychiatric stress, and accelerated aging. Brain Behav Immun 2019; 80:193-203. [PMID: 30872092 PMCID: PMC6660403 DOI: 10.1016/j.bbi.2019.03.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/27/2019] [Accepted: 03/09/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Longevity gene klotho (KL) is associated with age-related phenotypes but has not been evaluated against a direct human biomarker of cellular aging. We examined KL and psychiatric stress, including posttraumatic stress disorder (PTSD), which is thought to potentiate accelerated aging, in association with biomarkers of cellular aging. METHODS The sample comprised 309 white, non-Hispanic genotyped veterans with measures of epigenetic age (DNA methylation age), telomere length (n = 252), inflammation (C-reactive protein), psychiatric symptoms, metabolic function, and white matter neural integrity (diffusion tensor imaging; n = 185). Genotyping and DNA methylation were obtained on epi/genome-wide beadchips. RESULTS In gene by environment analyses, two KL variants (rs9315202 and rs9563121) interacted with PTSD severity (peak corrected p = 0.044) and sleep disturbance (peak corrected p = 0.034) to predict advanced epigenetic age. KL variant, rs398655, interacted with self-reported pain in association with slowed epigenetic age (corrected p = 0.048). A well-studied protective variant, rs9527025, was associated with slowed epigenetic age (p = 0.046). The peak PTSD interaction term (with rs9315202) also predicted C-reactive protein (p = 0.049), and white matter microstructural integrity in two tracts (corrected ps = 0.005 - 0.035). This SNP evidenced a main effect with an index of metabolic syndrome severity (p = 0.015). Effects were generally accentuated in older subjects. CONCLUSIONS Rs9315202 predicted multiple biomarkers of cellular aging such that psychiatric stress was more strongly associated with cellular aging in those with the minor allele. KL genotype may contribute to a synchronized pathological aging response to stress and could be a therapeutic target to alter the pace of cellular aging.
Collapse
|
54
|
Mazucanti CH, Kawamoto EM, Mattson MP, Scavone C, Camandola S. Activity-dependent neuronal Klotho enhances astrocytic aerobic glycolysis. J Cereb Blood Flow Metab 2019; 39:1544-1556. [PMID: 29493420 PMCID: PMC6681535 DOI: 10.1177/0271678x18762700] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mutations of the β-glucuronidase protein α-Klotho have been associated with premature aging, and altered cognitive function. Although highly expressed in specific areas of the brain, Klotho functions in the central nervous system remain unknown. Here, we show that cultured hippocampal neurons respond to insulin and glutamate stimulation by elevating Klotho protein levels. Conversely, AMPA and NMDA antagonism suppress neuronal Klotho expression. We also provide evidence that soluble Klotho enhances astrocytic aerobic glycolysis by hindering pyruvate metabolism through the mitochondria, and stimulating its processing by lactate dehydrogenase. Pharmacological inhibition of FGFR1, Erk phosphorylation, and monocarboxylic acid transporters prevents Klotho-induced lactate release from astrocytes. Taken together, these data suggest Klotho is a potential new player in the metabolic coupling between neurons and astrocytes. Neuronal glutamatergic activity and insulin modulation elicit Klotho release, which in turn stimulates astrocytic lactate formation and release. Lactate can then be used by neurons and other cells types as a metabolic substrate.
Collapse
Affiliation(s)
- Caio H Mazucanti
- 1 Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Elisa M Kawamoto
- 1 Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mark P Mattson
- 2 Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA.,3 Department of Neurosciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Cristoforo Scavone
- 1 Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Simonetta Camandola
- 2 Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA
| |
Collapse
|
55
|
Göttle P, Förster M, Weyers V, Küry P, Rejdak K, Hartung HP, Kremer D. An unmet clinical need: roads to remyelination in MS. Neurol Res Pract 2019; 1:21. [PMID: 33324887 PMCID: PMC7650135 DOI: 10.1186/s42466-019-0026-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/14/2019] [Indexed: 12/15/2022] Open
Abstract
Background In the central nervous system (CNS) myelin sheaths stabilize, protect, and electrically insulate axons. However, in demyelinating autoimmune CNS diseases such as multiple sclerosis (MS) these sheaths are destroyed which ultimately leads to neurodegeneration. The currently available immunomodulatory drugs for MS effectively control the (auto)inflammatory facets of the disease but are unable to regenerate myelin by stimulating remyelination via oligodendroglial precursor cells (OPCs). Accordingly, there is broad consensus that the implementation of new regenerative approaches constitutes the prime goal for future MS pharmacotherapy. Main text Of note, recent years have seen several promising clinical studies investigating the potential of substances and monoclonal antibodies such as, for instance, clemastine, opicinumab, biotin, simvastatin, quetiapin and anti-GNbAC1. However, beyond these agents which have often been re-purposed from other medical indications there is a multitude of further molecules influencing OPC homeostasis. Here, we therefore discuss these possibly beneficial regulators of OPC differentiation and assess their potential as new pharmacological targets for myelin repair in MS. Conclusion Remyelination remains the most important therapeutic treatment goal in MS in order to improve clinical deficits and to avert neurodegeneration. The promising molecules presented in this review have the potential to promote remyelination and therefore warrant further translational and clinical research.
Collapse
Affiliation(s)
- Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Moritz Förster
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Vivien Weyers
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Konrad Rejdak
- Department of Neurology, Medical University of Lublin, Lublin, Poland
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - David Kremer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| |
Collapse
|
56
|
Klotho Is Neuroprotective in the Superoxide Dismutase (SOD1 G93A) Mouse Model of ALS. J Mol Neurosci 2019; 69:264-285. [PMID: 31250273 DOI: 10.1007/s12031-019-01356-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 06/07/2019] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by the loss of motor neurons in the brain and spinal cord. ALS neuropathology is associated with increased oxidative stress, excitotoxicity, and inflammation. We and others reported that the anti-aging and cognition-enhancing protein Klotho is a neuroprotective, antioxidative, anti-inflammatory, and promyelinating protein. In mice, its absence leads to an extremely shortened life span and to multiple phenotypes resembling human aging, including motor and hippocampal neurodegeneration and cognitive impairment. In contrast, its overexpression extends life span, enhances cognition, and confers resistance against oxidative stress; it also reduces premature mortality and cognitive and behavioral abnormalities in an animal model for Alzheimer's disease (AD). These pleiotropic beneficial properties of Klotho suggest that Klotho could be a potent therapeutic target for preventing neurodegeneration in ALS. Klotho overexpression in the SOD1 mouse model of ALS resulted in delayed onset and progression of the disease and extended survival that was more prominent in females than in males. Klotho reduced the expression of neuroinflammatory markers and prevented neuronal loss with the more profound effect in the spinal cord than in the motor cortex. The effect of Klotho was accompanied by reduced expression of proinflammatory cytokines and enhanced the expression of antioxidative and promyelinating factors in the motor cortex and spinal cord of Klotho × SOD1 compared to SOD1 mice. Our study provides evidence that increased levels of Klotho alleviate ALS-associated pathology in the SOD1 mouse model and may serve as a basis for developing Klotho-based therapeutic strategies for ALS.
Collapse
|
57
|
Peshes-Yeloz N, Ungar L, Wohl A, Jacoby E, Fisher T, Leitner M, Nass D, Rubinek T, Wolf I, Cohen ZR. Role of Klotho Protein in Tumor Genesis, Cancer Progression, and Prognosis in Patients with High-Grade Glioma. World Neurosurg 2019; 130:e324-e332. [PMID: 31228703 DOI: 10.1016/j.wneu.2019.06.082] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND Klotho, a single-pass transmembrane protein associated with premature aging, acts as a tumor suppressor gene by inhibiting insulin/insulin-like growth factor-1 and fibroblast growth factor pathways. Downregulated Klotho expression is reported in melanoma, mesothelioma, bladder, breast, gastric, cervix, lung, and kidney cancers and is associated with a poor prognosis. Klotho expression and Klotho promoter hypermethylation are predictive factors for patient prognosis. METHODS To investigate the potential role of Klotho in glioblastoma-multiforme (GBM), 22 GBM samples were collected from the Sheba Tumor Bank and examined. RESULTS We found that increased Klotho messenger ribonucleic acid (RNA) expression predicted longer survival (P = 0.03) of GBM patients. Methylation analysis was performed on bisulfite-treated deoxyribonucleic acid from the GBM patient samples using ionization time-of-flight mass spectrometry according to the Sequenom EpiTYPER protocols. Klotho promoter hypermethylation was detected in 65% of the GBM samples and correlated significantly with improved survival (P < 0.04). We found 3 major Klotho promotor hypermethylation sites located 585-579 bp, 540-533 bp, and 537-534 bp upstream of the transcription start site. Methylated deoxyribonucleic acid immunoprecipitation studies confirmed these results. Notably, the messenger RNA expression in these GBM samples revealed an unexpected linear correlation with methylation of these 3 hypermethylation sites identified in the Klotho promotor. Thus Klotho expression and methylation could predict prognosis in patients with GBM. CONCLUSIONS Epigenetic regulation in GBM appears to be complicated. Specific CpG islands affect genes or micro RNAs that interact to control Klotho expression. The diverse effects of these islands may be due to unique factors of GBM.
Collapse
Affiliation(s)
- Naama Peshes-Yeloz
- Department of Neurosurgery, Sheba Medical Center, Ramat Gan; Affiliated with the Sackler School of Medicine, Tel Aviv, Israel
| | - Lior Ungar
- Department of Neurosurgery, Sheba Medical Center, Ramat Gan; Affiliated with the Sackler School of Medicine, Tel Aviv, Israel
| | - Anton Wohl
- Department of Neurosurgery, Sheba Medical Center, Ramat Gan; Affiliated with the Sackler School of Medicine, Tel Aviv, Israel
| | - Elad Jacoby
- Cancer Research Center, Sheba Medical Center, Ramat Gan; Affiliated with the Sackler School of Medicine, Tel Aviv, Israel
| | - Tamar Fisher
- Cancer Research Center, Sheba Medical Center, Ramat Gan; Affiliated with the Sackler School of Medicine, Tel Aviv, Israel
| | - Moshe Leitner
- Cancer Research Center, Sheba Medical Center, Ramat Gan; Affiliated with the Sackler School of Medicine, Tel Aviv, Israel
| | - Dvora Nass
- Institute of Pathology, Sheba Medical Center, Ramat Gan; Affiliated with the Sackler School of Medicine, Tel Aviv, Israel
| | - Tamar Rubinek
- Institute of Oncology, Tel-Aviv Medical Center, Tel-Aviv; Affiliated with the Sackler School of Medicine, Tel Aviv, Israel
| | - Ido Wolf
- Institute of Oncology, Tel-Aviv Medical Center, Tel-Aviv; Affiliated with the Sackler School of Medicine, Tel Aviv, Israel
| | - Zvi R Cohen
- Department of Neurosurgery, Sheba Medical Center, Ramat Gan; Affiliated with the Sackler School of Medicine, Tel Aviv, Israel.
| |
Collapse
|
58
|
Li D, Jing D, Liu Z, Chen Y, Huang F, Behnisch T. Enhanced Expression of Secreted α-Klotho in the Hippocampus Alters Nesting Behavior and Memory Formation in Mice. Front Cell Neurosci 2019; 13:133. [PMID: 31001090 PMCID: PMC6454015 DOI: 10.3389/fncel.2019.00133] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/18/2019] [Indexed: 12/30/2022] Open
Abstract
The klotho gene family consists of α-, β-, and γ-Klotho, which encode type I single-pass transmembrane proteins with large extracellular domains. α-Klotho exists as a full-length membrane-bound and as a soluble form after cleavage of the extracellular domain. Due to gene splicing, a short extracellular Klotho form can be expressed and secreted. Inactivation of α-Klotho leads to a phenotype that resembles accelerated aging, as the expression level of the α-Klotho protein in the hippocampal formation of mice decreases with age. Here, we show that intrahippocampal viral expression of secreted human α-Klotho alters social behavior and memory formation. Interestingly, overexpression of secreted human α-Klotho in the CA1 changed the nest-building behavior and improved object recognition, object location and passive avoidance memory. Moreover, α-Klotho overexpression increased hippocampal synaptic transmission in response to standardized stimulation strengths, altered paired-pulse facilitation of synaptic transmission, and enhanced activity-dependent synaptic plasticity. These results indicate that memory formation benefits from an augmented level of secreted α-Klotho.
Collapse
Affiliation(s)
- Dongxue Li
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Dongqing Jing
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ziyang Liu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ying Chen
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Fang Huang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Thomas Behnisch
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
59
|
FGF-23 Deficiency Impairs Hippocampal-Dependent Cognitive Function. eNeuro 2019; 6:eN-NRS-0469-18. [PMID: 30911673 PMCID: PMC6430630 DOI: 10.1523/eneuro.0469-18.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 01/05/2023] Open
Abstract
Fibroblast growth factor receptor (FGFR) and α-Klotho transduce FGF-23 signaling in renal tubules to maintain systemic phosphate/vitamin D homeostasis. Mice deficient for either the ligand, FGF-23, or the co-receptor, Klotho, are phenocopies with both showing rapid and premature development of multiple aging-like abnormalities. Such similarity in phenotype, suggests that FGF-23 and Klotho have co-dependent systemic functions. Recent reports revealed inverse central nervous system (CNS) effects of Klotho deficiency or Klotho overexpression on hippocampal synaptic, neurogenic, and cognitive functions. However, it is unknown whether FGF-23 deficiency effects function of the hippocampus. We report that, similar to Klotho-deficient mice, FGF-23-deficient mice develop dose-dependent, hippocampal-dependent cognitive impairment. However, FGF-23-deficient brains had no gross structural or developmental defects, no change in hippocampal synaptic plasticity, and only minor impairment to postnatal hippocampal neurogenesis. Together, these data provide evidence that FGF-23 deficiency impairs hippocampal-dependent cognition but otherwise results in a brain phenotype that is distinct from the KL-deficient mouse.
Collapse
|
60
|
Prokhorova TA, Boksha IS, Savushkina OK, Tereshkina EB, Burbaeva GS. [α-Klotho protein in neurodegenerative and mental diseases]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:80-88. [PMID: 30778037 DOI: 10.17116/jnevro201911901180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The review aims to attract attention of psychiatrists and neurologists to a role of α-Klotho protein in biochemical mechanisms that counteract pathogenic processes of neurodegenerative and psychiatric diseases and to possible therapeutic potential of the protein. Basing on the analysis of contemporary literature, the authors summarized the results of model experiments and a few clinical trials (in psychiatry and neurology) indicating the role of α-Klotho protein in the brain processes of neurogenesis, dendrite growth, myelination (oligodendroglia differentiation and activity), regulation of antioxidant system, and synthesis of glutamate neurotransmitter system components, regulation of the activity and synthesis of ion channel protein components and membrane transporters, synaptic plasticity. It is concluded that α-Klotho protein can be used for therapeutic purposes in diseases associated with pathological brain aging, and/or in diseases associated with insufficient synthesis of this protein.
Collapse
Affiliation(s)
| | - I S Boksha
- Mental Health Research Centre, Moscow, Russia
| | | | | | | |
Collapse
|
61
|
Lentiviral vector-mediated overexpression of Klotho in the brain improves Alzheimer's disease-like pathology and cognitive deficits in mice. Neurobiol Aging 2019; 78:18-28. [PMID: 30851437 DOI: 10.1016/j.neurobiolaging.2019.02.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/25/2019] [Accepted: 02/02/2019] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the most common type of senile dementia. The antiaging gene Klotho is reported to decline in the brain of patients and animals with AD. However, the role of Klotho in the progression of AD remains elusive. The present study explored the effects and underlying mechanism of Klotho in a mouse model of AD. The upregulation of cerebral Klotho expression was mediated by an intracerebroventricular injection of a lentiviral vector that encoded Klotho (LV-KL) in 7-month-old amyloid precursor protein/presenilin 1 transgenic mice. Three months later, LV-KL significantly induced Klotho overexpression in the brain and effectively ameliorated cognitive deficit and AD-like pathology in amyloid precursor protein/presenilin 1 mice. LV-KL induced autophagy activation and protein kinase B/mammalian target of rapamycin inhibition both in AD mice and BV2 murine microglia. These results suggest that the upregulation of Klotho expression in the brain may promote the autophagic clearance of amyloid beta and protect against cognitive deficits in AD mice. These findings highlight the preventive and therapeutic potential of Klotho for the treatment of AD.
Collapse
|
62
|
Šťastník O, Mrkvicová E, Pavlata L, Roztočilová A, Jůzl M, Pytel R, Vyhnánek T, Martinek P. Influence of feeding colored wheat varieties on selected quality parameters of broiler chicken”™s meat. POTRAVINARSTVO 2018. [DOI: 10.5219/986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The effect of feeding colored wheat varieties (PS Karkulka and Skorpion) on chicken´s performance and texture, color and sensory characteristics of broiler chicken´s meat were evaluated in this study. The experiment was performed with 66 of Ross 308 cockerels. Cockerels were divided into three equal groups. The two experimental groups received feed mixtures containing 40% of wheats with different grain colour: groups PS Karkulka (n = 22) with PS Karkulka wheat cultivar and Skorpion (n = 22) with Skorpion cultivar. The third group (n = 22) had 40% of common wheat Vánek cultivar (Control). The live weight of broilers between all three groups was not significant different, as well as carcass yield and chemical composition of breast and thigh meat of chickens. In the parameter Razor Shear Force was found statistically significant higher breast meat tenderness in PS Karkulka against Control and Skorpion groups. In parameters L* and b* of colour of the meat samples was found statistically significant higher value in L* parameter in Skorpion group and b* parameter was higher in Control group. The total colour change was 2.25 and 2.53 for PS Karkulka and Skorpion group, respectively. In sensory analysis of broilers breast muscle was found statistically significant differences in odour, colour, fibreness, chewiness, juiciness and flavour parameter. The fatty taste parameter was non-significant. The odour parameter of chicken´s breast muscle was significantly lower in Skorpion group against PS Karkulka and Control groups. The significantly most intense colour of breast muscle was found in Control group versus Skorpion and PS Karkulka groups (91.71 mm, 79.71 mm and 71.15 mm, respectively). The fibreness parameter was significantly higher for Control group, as well. Significantly higher chewiness of breast meat was in Control (68.49 mm) than PS Karkulka (52.02 mm) and Skorpion (43.32 mm) group. The feeding of wheat cultivars with different grain pigmentation had no effect on performance parameters of broiler chicken´s as well as to it´s body and chemical composition of breast and thigh meat in this study.
Collapse
|
63
|
Secreted αKlotho isoform protects against age-dependent memory deficits. Mol Psychiatry 2018; 23:1937-1947. [PMID: 29086766 DOI: 10.1038/mp.2017.211] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/24/2017] [Accepted: 07/28/2017] [Indexed: 11/08/2022]
Abstract
αKlotho is a gene regulator of aging, increasing life expectancy when overexpressed and accelerating the development of aging phenotypes when inhibited. In mice, expression levels of the secreted isoform Klotho (s-KL) are very high in the brain, suggesting that s-KL activity may have an important role in the nervous system. Here we study the functional relevance at behavioural level of modifying s-KL levels in the aging brain. We used AAVrh10 vectors to deliver and sustained expression of s-KL in 6- and 12-month-old wild-type C57BL/6J males. This study demonstrates for we believe the first time in vivo that 6 months after a single injection of s-KL into the central nervous system, long-lasting and quantifiable enhancement of learning and memory capabilities are found. More importantly, cognitive improvement is also observable in 18-month-old mice treated once, at 12 months of age. These findings demonstrate the therapeutic potential of s-KL as a treatment for cognitive decline associated with aging.
Collapse
|
64
|
Mansoor SR, Hashemian M, Khalili-Fomeshi M, Ashrafpour M, Moghadamnia AA, Ghasemi-Kasman M. Upregulation of klotho and erythropoietin contributes to the neuroprotection induced by curcumin-loaded nanoparticles in experimental model of chronic epilepsy. Brain Res Bull 2018; 142:281-288. [PMID: 30130550 DOI: 10.1016/j.brainresbull.2018.08.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 07/26/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022]
Abstract
Klotho, which is a life extension factor, and erythropoietin (EPO) have been introduced as effective neuroprotective factors in several neurological disorders. The present study is an attempt to examine the potential role of klotho and EPO in therapeutic effect of curcumin-loaded nanoparticles (NPs) in pentylenetetrazol (PTZ)-induced kindling model. In order to induce the kindling model, PTZ was administrated intraperitoneally (i.p.) at dose of 36.5 mg/kg every other day for 20 days. Male NMRI mice received pre-treatment of free curcumin or curcumin-loaded NPs (12.5 mg/kg, i.p.) 10 days before PTZ injection and this was continued until 1 h before each PTZ injection. Immunostaining against NeuN, as a marker of neuronal maturation, and EPO was performed on hippocampal brain sections. Quantitative real time polymerase chain reaction (qRT-PCR) was conducted to assess the expression levels of tumor necrosis factor-α (TNF-α), klotho and EPO in the hippocampus. Immunostaining data indicated that treatment with curcumin-loaded NPs significantly alleviates the neuronal cell death in PTZ receiving animals. Curcumin-loaded NPs effectively upregulated the levels of EPO and klotho in PTZ receiving animals. Furthermore, mRNA level of TNF-α was considerably reduced in animals undergone curcumin-loaded NPs treatment. Overall, the results of this study suggest that downregulation of TNF-α and consequent upregulation of klotho and EPO might contribute to the neuroprotective effect of curcumin-loaded NPs in experimental model of epilepsy.
Collapse
Affiliation(s)
| | - Mona Hashemian
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | - Manouchehr Ashrafpour
- Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Physiology, Faculty of Medical Sciences, Babol University of Medical Sciences, Babol, Iran
| | - Ali Akbar Moghadamnia
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Pharmacology, Faculty of Medical Sciences, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
65
|
Abstract
Brain expression of klotho was first described with the initial discovery of the klotho gene. The prominent age-regulating effects of klotho are attributed to regulation of ion homeostasis through klotho function in the kidney. However, recent advances identified brain functions and cell populations, including adult hippocampal neural progenitors, which require klotho. As well, both human correlational studies and mouse models of disease show that klotho is protective against multiple neurological and psychological disorders. This review focuses on current knowledge as to how the klotho protein effects the brain.
Collapse
Affiliation(s)
- Hai T Vo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ann M Laszczyk
- Department of Cell and Developmental Biology, University of Michigan, Zina Pitcher Pl, Ann Arbor, MI, USA
| | - Gwendalyn D King
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
66
|
Torbus-Paluszczak M, Bartman W, Adamczyk-Sowa M. Klotho protein in neurodegenerative disorders. Neurol Sci 2018; 39:1677-1682. [PMID: 30062646 PMCID: PMC6154120 DOI: 10.1007/s10072-018-3496-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 07/05/2018] [Indexed: 11/28/2022]
Abstract
The Klotho protein is a recently discovered protein and its overexpression is associated with life extension. Klotho deficiency or silencing of the Klotho gene in mice leads to an accelerated aging and short life, whereas overexpression of Klotho in mice extends lifespan. Klotho participates in many metabolic pathways and is highly expressed in the kidneys, the choroid plexus and neurons. It plays a key role in the calcium-phosphate metabolism, remyelination, and cognitive processes. The present paper is a short review of the literature on the role of Klotho in neurodegenerative disorders, with special attention paid to multiple sclerosis. The neuroprotective function of Klotho is also reported. It is also important to consider potential clinical applications of Klotho that might be useful in the treatment of many diseases.
Collapse
Affiliation(s)
- Magdalena Torbus-Paluszczak
- Department of Neurology, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Katowice, Poland, ul. 3-go Maja 13-15, 41-800, Zabrze, Poland.
| | - Wojciech Bartman
- Department of Neurology, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Katowice, Poland, ul. 3-go Maja 13-15, 41-800, Zabrze, Poland
| | - Monika Adamczyk-Sowa
- Department of Neurology, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Katowice, Poland, ul. 3-go Maja 13-15, 41-800, Zabrze, Poland
| |
Collapse
|
67
|
Stem Cells as Potential Targets of Polyphenols in Multiple Sclerosis and Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1483791. [PMID: 30112360 PMCID: PMC6077677 DOI: 10.1155/2018/1483791] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/19/2018] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) and multiple sclerosis are major neurodegenerative diseases, which are characterized by the accumulation of abnormal pathogenic proteins due to oxidative stress, mitochondrial dysfunction, impaired autophagy, and pathogens, leading to neurodegeneration and behavioral deficits. Herein, we reviewed the utility of plant polyphenols in regulating proliferation and differentiation of stem cells for inducing brain self-repair in AD and multiple sclerosis. Firstly, we discussed the genetic, physiological, and environmental factors involved in the pathophysiology of both the disorders. Next, we reviewed various stem cell therapies available and how they have proved useful in animal models of AD and multiple sclerosis. Lastly, we discussed how polyphenols utilize the potential of stem cells, either complementing their therapeutic effects or stimulating endogenous and exogenous neurogenesis, against these diseases. We suggest that polyphenols could be a potential candidate for stem cell therapy against neurodegenerative disorders.
Collapse
|
68
|
Anthocyanin bio-fortified colored wheat: Nutritional and functional characterization. PLoS One 2018; 13:e0194367. [PMID: 29617385 PMCID: PMC5884506 DOI: 10.1371/journal.pone.0194367] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 03/01/2018] [Indexed: 12/17/2022] Open
Abstract
Colored wheat, rich in anthocyanins, has created interest among the breeders and baking industry. This study was aimed at understanding the nutritional and product making potential of our advanced, high yielding and regionally adapted colored wheat lines. Our results indicated that our advanced colored wheat lines exhibited higher anthocyanin content and antioxidant activity than donor wheat lines and it varied in the order of white<purple<blue<black wheat. UPLC chromatogram revealed that anthocyanin composition and peak pattern is not only dependent on donor genotype but also background of recipient genotype. Interestingly, the purple wheat extract showed highest anti-inflammatory effect and followed the trend of white<blue<black<purple. Nutritional (carbohydrates, sugar, protein, ash, dietary fibre and vitamins) and processing parameters in relation to end-use quality (SDS sedimentation, gluten content, alveograph) of advanced colored lines were similar to high yielding white wheat cultivar. Colored wheat lines showed high iron and zinc content compared to white wheat indicating double bio-fortification. Therefore, our advanced colored wheat lines with high anthocyanin, iron and zinc contents showed antioxidant and anti-inflammatory activity and possessed desirable features for product making and commercial utilization.
Collapse
|
69
|
Emami Aleagha MS, Harirchian MH, Lavasani S, Javan M, Allameh A. Differential Expression of Klotho in the Brain and Spinal Cord is Associated with Total Antioxidant Capacity in Mice with Experimental Autoimmune Encephalomyelitis. J Mol Neurosci 2018. [DOI: 10.1007/s12031-018-1058-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
70
|
Morar B, Badcock JC, Phillips M, Almeida OP, Jablensky A. The longevity gene Klotho is differentially associated with cognition in subtypes of schizophrenia. Schizophr Res 2018; 193:348-353. [PMID: 28673754 DOI: 10.1016/j.schres.2017.06.054] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 06/22/2017] [Accepted: 06/27/2017] [Indexed: 01/10/2023]
Abstract
Cognitive impairment is a core feature of schizophrenia and impacts negatively the functioning of affected individuals. Cognitive decline correlates with aging, and is the primary cause of loss of independence and reduced quality of life. The klotho gene is a key modulator of aging, with expression deficiency resulting in premature aging, while overexpression extends lifespan and enhances cognition. A haplotype and functional human variant of the gene, KL-VS, increases expression and promotes longevity. KL-VS heterozygosity is associated with enhanced cognition and a larger volume of the right dorsolateral prefrontal cortex, a region involved in planning and decision-making, which is especially susceptible to shrinkage with age. We examined the effect of KL-VS heterozygosity on cognition in 497 schizophrenia patients and 316 healthy controls from the Western Australian Family Study of Schizophrenia (WAFSS) who had been comprehensively characterised by neurocognitive tests and classified into cognitively deficient (CD) and cognitively "spared" (CS) clusters. An older, cognitively normal population sample from the Health in Men Study (HIMS) was included to allow assessment of heterozygosity and memory in aged individuals. We show that heterozygosity is associated with better learning and memory in the younger WAFSS healthy controls but not in the aging HIMS sample. However, in schizophrenia patients, KL-VS has a selective effect on memory, with heterozygotes in CD and CS clusters performing worse than non-carriers. This effect was significant and more severe in the CD cluster, reinforcing the utility of subtyping patients into CD and CS clusters that may differ in their genetic underpinnings.
Collapse
Affiliation(s)
- Bharti Morar
- Centre for Clinical Research in Neuropsychiatry, School of Psychiatry and Clinical Neurosciences, University of Western Australia, MRF Building, 50 Murray Street, Perth 6000, Australia; Cooperative Research Centre for Mental Health, Carlton South, Victoria, Australia; Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, 6 Verdun Street, Nedlands, WA 6009, Australia.
| | - Johanna C Badcock
- Centre for Clinical Research in Neuropsychiatry, School of Psychiatry and Clinical Neurosciences, University of Western Australia, MRF Building, 50 Murray Street, Perth 6000, Australia; Cooperative Research Centre for Mental Health, Carlton South, Victoria, Australia
| | - Michael Phillips
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, 6 Verdun Street, Nedlands, WA 6009, Australia
| | - Osvaldo P Almeida
- WA Centre for Health and Ageing, Centre for Medical Research, Perth, Australia
| | - Assen Jablensky
- Centre for Clinical Research in Neuropsychiatry, School of Psychiatry and Clinical Neurosciences, University of Western Australia, MRF Building, 50 Murray Street, Perth 6000, Australia; Cooperative Research Centre for Mental Health, Carlton South, Victoria, Australia
| |
Collapse
|
71
|
Robinson AA, Abraham CR, Rosene DL. Candidate molecular pathways of white matter vulnerability in the brain of normal aging rhesus monkeys. GeroScience 2018; 40:31-47. [PMID: 29357021 PMCID: PMC5832663 DOI: 10.1007/s11357-018-0006-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 01/08/2018] [Indexed: 12/20/2022] Open
Abstract
Mammalian aging is associated with decline in cognitive functions. Studies searching for a cause of cognitive aging initially focused on neuronal loss but quantitative investigations of rat, monkey, and human brain using stereology demonstrated that in normal aging, unlike in neurodegenerative disease, neurons are not lost. Instead, electron microscopic and MRI studies in normal aging monkeys revealed age-related damage to myelin sheaths, loss of axons, and reduction in white matter volume which correlates with cognitive impairments. However, little is known about the cause of myelin defects or associated axon loss. The present study investigates the effect of age on signaling pathways between oligodendroglia and neurons using a custom PCR array to assess the expression of 87 genes of interest in cortical gray matter and white matter from the inferior parietal lobe (IPL) of normal rhesus monkeys ranging in age from 4.2 to 30.4 years old. From this array data, five target genes of interest were selected for further analysis to confirm gene expression and measure protein expression. The most interesting target gene identified is brain-derived neurotrophic factor (BDNF), which was the only gene that was altered at both mRNA and protein levels. In gray matter, BDNF mRNA was decreased. While the level of the mature form of the protein was unchanged, there was a specific decrease in the precursor form of BDNF. These alterations in the BDNF in gray matter could contribute to the vulnerability and loss of the axons with age.
Collapse
Affiliation(s)
- Amy A. Robinson
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118 USA
| | - Carmela R. Abraham
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118 USA
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118 USA
| | - Douglas L. Rosene
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118 USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322 USA
| |
Collapse
|
72
|
Chen CD, Zeldich E, Li Y, Yuste A, Abraham CR. Activation of the Anti-Aging and Cognition-Enhancing Gene Klotho by CRISPR-dCas9 Transcriptional Effector Complex. J Mol Neurosci 2018; 64:175-184. [PMID: 29352444 DOI: 10.1007/s12031-017-1011-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 11/28/2017] [Indexed: 12/13/2022]
Abstract
Multiple lines of evidence show that the anti-aging and cognition-enhancing protein Klotho fosters neuronal survival, increases the anti-oxidative stress defense, and promotes remyelination of demyelinated axons. Thus, upregulation of the Klotho gene can potentially alleviate the symptoms and/or prevent the progression of age-associated neurodegenerative diseases such as Alzheimer's disease and demyelinating diseases such as multiple sclerosis. Here we used a CRISPR-dCas9 complex to investigate single-guide RNA (sgRNA) targeting the Klotho promoter region for efficient transcriptional activation of the Klotho gene. We tested the sgRNAs within the - 1 to - 300 bp of the Klotho promoter region and identified two sgRNAs that can effectively enhance Klotho gene transcription. We examined the transcriptional activation of the Klotho gene using three different systems: a Firefly luciferase (FLuc) and NanoLuc luciferase (NLuc) coincidence reporter system, a NLuc knock-in in Klotho 3'-UTR using CRISPR genomic editing, and two human cell lines: neuronal SY5Y cells and kidney HK-2 cells that express Klotho endogenously. The two sgRNAs enhanced Klotho expression at both the gene and protein levels. Our results show the feasibility of gene therapy for targeting Klotho using CRISPR technology. Enhancing Klotho levels has a therapeutic potential for increasing cognition and treating age-associated neurodegenerative, demyelinating and other diseases, such as chronic kidney disease and cancer.
Collapse
Affiliation(s)
- Ci-Di Chen
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.,Klogene Therapeutics, Inc., Boston, MA, USA
| | - Ella Zeldich
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.,Klogene Therapeutics, Inc., Boston, MA, USA
| | - Yuexuan Li
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | | | - Carmela R Abraham
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA. .,Klogene Therapeutics, Inc., Boston, MA, USA. .,Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
73
|
Uncoupling N-acetylaspartate from brain pathology: implications for Canavan disease gene therapy. Acta Neuropathol 2018; 135:95-113. [PMID: 29116375 PMCID: PMC5756261 DOI: 10.1007/s00401-017-1784-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 11/25/2022]
Abstract
N-Acetylaspartate (NAA) is the second most abundant organic metabolite in the brain, but its physiological significance remains enigmatic. Toxic NAA accumulation appears to be the key factor for neurological decline in Canavan disease—a fatal neurometabolic disorder caused by deficiency in the NAA-degrading enzyme aspartoacylase. To date clinical outcome of gene replacement therapy for this spongiform leukodystrophy has not met expectations. To identify the target tissue and cells for maximum anticipated treatment benefit, we employed comprehensive phenotyping of novel mouse models to assess cell type-specific consequences of NAA depletion or elevation. We show that NAA-deficiency causes neurological deficits affecting unconscious defensive reactions aimed at protecting the body from external threat. This finding suggests, while NAA reduction is pivotal to treat Canavan disease, abrogating NAA synthesis should be avoided. At the other end of the spectrum, while predicting pathological severity in Canavan disease mice, increased brain NAA levels are not neurotoxic per se. In fact, in transgenic mice overexpressing the NAA synthesising enzyme Nat8l in neurons, supra-physiological NAA levels were uncoupled from neurological deficits. In contrast, elimination of aspartoacylase expression exclusively in oligodendrocytes elicited Canavan disease like pathology. Although conditional aspartoacylase deletion in oligodendrocytes abolished expression in the entire CNS, the remaining aspartoacylase in peripheral organs was sufficient to lower NAA levels, delay disease onset and ameliorate histopathology. However, comparable endpoints of the conditional and complete aspartoacylase knockout indicate that optimal Canavan disease gene replacement therapies should restore aspartoacylase expression in oligodendrocytes. On the basis of these findings we executed an ASPA gene replacement therapy targeting oligodendrocytes in Canavan disease mice resulting in reversal of pre-existing CNS pathology and lasting neurological benefits. This finding signifies the first successful post-symptomatic treatment of a white matter disorder using an adeno-associated virus vector tailored towards oligodendroglial-restricted transgene expression.
Collapse
|
74
|
Gadelkareem RA, Moeen AM, Reda A, Azoz NM, Elhadad AF, Taha TM, Mohammed N, Taha MI. Experience of a Tertiary-Level Urology Center in the Clinical Urological Events of Rare and Very Rare Incidence. III. Psychourological Events: 1. Psychic Anuria. Urol Int 2018; 101:80-84. [PMID: 29843132 DOI: 10.1159/000488881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/28/2018] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Psychic anuria is an old term, referring to a very rare psycho-urological event that has scarcely been studied so far. MATERIALS AND METHODS A retrospective study of the patients with psychic anuria presented to Assiut Urology and Nephrology Hospital during the period July 1991-June 2016 was done. Psychic anuria was defined, and the demographic and clinical characteristics including the methods of diagnosis and management were studied. RESULTS Of more than 3,800 cases of anuria, 9 female patients (0.24%) experienced psychic anuria in the age range of 17-43 years. Cardinal clinical findings included anuria for 36-72 h with absence of organic causes and normal renal function tests. Psychosocial risk factors were reported in the 9 cases. Anuria was documented by reliable history (56%) or observable urine collection (44%). Diagnosis was done by exclusion, where the investigations revealed no organic causes. Seven cases responded to the placebo intervention and 2 cases were self-limiting and resolved spontaneously. CONCLUSIONS Psychic anuria is an extremely rare urological emergency that presents, mainly, in young adult females with unknown mechanisms. Renal vasoconstriction following psychosocial stressors is suggested. It is diagnosed by exclusion and resolves spontaneously or responds to placebo intervention as a mental distraction technique.
Collapse
Affiliation(s)
- Rabea A Gadelkareem
- Assiut Urology and Nephrology Hospital, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed M Moeen
- Assiut Urology and Nephrology Hospital, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed Reda
- Assiut Urology and Nephrology Hospital, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Nashwa M Azoz
- Department of Internal Medicine-Nephrology Unit, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ali F Elhadad
- Department of Neuropsychiatry, Faculty of Medicine, Al-Azhar University-Assiut Branch, Assiut, Egypt
| | - Taha M Taha
- Assiut Urology and Nephrology Hospital, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Nasreldin Mohammed
- Assiut Urology and Nephrology Hospital, Faculty of Medicine, Assiut University, Assiut, Egypt
- Department of Urology, Martin-Luther University, Halle, Germany
| | - Mohammed I Taha
- Assiut Urology and Nephrology Hospital, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
75
|
Richter B, Faul C. FGF23 Actions on Target Tissues-With and Without Klotho. Front Endocrinol (Lausanne) 2018; 9:189. [PMID: 29770125 PMCID: PMC5940753 DOI: 10.3389/fendo.2018.00189] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/06/2018] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor (FGF) 23 is a phosphaturic hormone whose physiologic actions on target tissues are mediated by FGF receptors (FGFR) and klotho, which functions as a co-receptor that increases the binding affinity of FGF23 for FGFRs. By stimulating FGFR/klotho complexes in the kidney and parathyroid gland, FGF23 reduces renal phosphate uptake and secretion of parathyroid hormone, respectively, thereby acting as a key regulator of phosphate metabolism. Recently, it has been shown that FGF23 can also target cell types that lack klotho. This unconventional signaling event occurs in an FGFR-dependent manner, but involves other downstream signaling pathways than in "classic" klotho-expressing target organs. It appears that klotho-independent signaling mechanisms are only activated in the presence of high FGF23 concentrations and result in pathologic cellular changes. Therefore, it has been postulated that massive elevations in circulating levels of FGF23, as found in patients with chronic kidney disease, contribute to associated pathologies by targeting cells and tissues that lack klotho. This includes the induction of cardiac hypertrophy and fibrosis, the elevation of inflammatory cytokine expression in the liver, and the inhibition of neutrophil recruitment. Here, we describe the signaling and cellular events that are caused by FGF23 in tissues lacking klotho, and we discuss FGF23's potential role as a hormone with widespread pathologic actions. Since the soluble form of klotho can function as a circulating co-receptor for FGF23, we also discuss the potential inhibitory effects of soluble klotho on FGF23-mediated signaling which might-at least partially-underlie the pleiotropic tissue-protective functions of klotho.
Collapse
|
76
|
Xuan NT, Hai NV. Changes in expression of klotho affect physiological processes, diseases, and cancer. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2018; 21:3-8. [PMID: 29372030 PMCID: PMC5776433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Klotho (KL) encodes a single-pass transmembrane protein and is predominantly expressed in the kidney, parathyroid glands, and choroid plexus. Genetic studies on the KL gene have revealed that DNA hypermethylation is one of the major risk factors for aging, diseases, and cancer. Besides, KL exerts anti-inflammatory and anti-tumor effects by regulating signaling pathways and the expression of target genes. KL participates in modulation of the insulin/insulin-like growth factor-1 (IGF-1) signaling, which induces the growth hormone (GH) secretion. Accordingly, KL mutant mice display multiple aging-like phenotypes, which are ameliorated by overexpression of KL. Therefore, KL is an important contributor to lifespan. KL is further identified as a regulator of calcium (Ca2+) channel-dependent cell physiological processes. KL has been also shown to induce cancer cell apoptosis, thus, it is considered as a potential tumor suppressor. Our recent studies have indicated that KL modulates an influx of Ca2+ from the extracellular space, leading to a change in CCL21-dependent migration in dendritic cells (DCs). Interestingly, the regulation of the expression of KL was mediated through a phosphoinositide 3-kinase (PI3K) pathway in DCs. Moreover, downregulating of KL expression by using siRNA knockdown technique, we observed that the expression of Ca2+ channels including Orai3, but not Orai1, Orai2, TRPV5 and TRPV6 was significantly reduced in KL-silenced as compared to control BMDCs. Clearly, additional research is required to define the role of KL in the regulation of organismic and cellular functions through the PI3K signaling and the expression of the Ca2+ channels.
Collapse
Affiliation(s)
- Nguyen Thi Xuan
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam,Corresponding author: Nguyen Thi Xuan. Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam. Tel: +84-24-32121143; E-mail:
| | - Nong Van Hai
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| |
Collapse
|
77
|
Edo Á, Espinosa-Parrilla JF. Soluble interleukin 23 receptor gene therapy with adeno-associated vectors for the treatment of multiple sclerosis. Neural Regen Res 2017; 12:1605-1606. [PMID: 29171419 PMCID: PMC5696835 DOI: 10.4103/1673-5374.217327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Ángel Edo
- Gene Therapy for Central Nervous System Group, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Juan Francisco Espinosa-Parrilla
- Gene Therapy for Central Nervous System Group, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
78
|
Choi CI, Yoo KH, Hussaini SMQ, Jeon BT, Welby J, Gan H, Scarisbrick IA, Zhang Z, Baker DJ, van Deursen JM, Rodriguez M, Jang MH. The progeroid gene BubR1 regulates axon myelination and motor function. Aging (Albany NY) 2017; 8:2667-2688. [PMID: 27922816 PMCID: PMC5191862 DOI: 10.18632/aging.101032] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 08/25/2016] [Indexed: 01/22/2023]
Abstract
Myelination, the process by which oligodendrocytes form the myelin sheath around axons, is key to axonal signal transduction and related motor function in the central nervous system (CNS). Aging is characterized by degenerative changes in the myelin sheath, although the molecular underpinnings of normal and aberrant myelination remain incompletely understood. Here we report that axon myelination and related motor function are dependent on BubR1, a mitotic checkpoint protein that has been linked to progeroid phenotypes when expressed at low levels and healthy lifespan when overabundant. We found that oligodendrocyte progenitor cell proliferation and oligodendrocyte density is markedly reduced in mutant mice with low amounts of BubR1 (BubR1H/H mice), causing axonal hypomyelination in both brain and spinal cord. Expression of essential myelin-related genes such as MBP and PLP1 was significantly reduced in these tissues. Consistent with defective myelination, BubR1H/H mice exhibited various motor deficits, including impaired motor strength, coordination, and balance, irregular gait patterns and reduced locomotor activity. Collectively, these data suggest that BubR1 is a key determinant of oligodendrocyte production and function and provide a molecular entry point to understand age-related degenerative changes in axon myelination.
Collapse
Affiliation(s)
- Chan-Il Choi
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Ki Hyun Yoo
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | - Byeong Tak Jeon
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - John Welby
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Haiyun Gan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Isobel A Scarisbrick
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Zhiguo Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Darren J Baker
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.,Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Jan M van Deursen
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.,Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Moses Rodriguez
- Departments of Neurology and Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Mi-Hyeon Jang
- Department of Neurologic Surgery, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.,Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
79
|
Laszczyk AM, Fox-Quick S, Vo HT, Nettles D, Pugh PC, Overstreet-Wadiche L, King GD. Klotho regulates postnatal neurogenesis and protects against age-related spatial memory loss. Neurobiol Aging 2017; 59:41-54. [PMID: 28837861 PMCID: PMC5612914 DOI: 10.1016/j.neurobiolaging.2017.07.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 06/22/2017] [Accepted: 07/21/2017] [Indexed: 12/29/2022]
Abstract
Although the absence of the age-regulating klotho protein causes klotho-deficient mice to rapidly develop cognitive impairment and increasing klotho enhances hippocampal-dependent memory, the cellular effects of klotho that mediate hippocampal-dependent memory function are unknown. Here, we show premature aging of the klotho-deficient hippocampal neurogenic niche as evidenced by reduced numbers of neural stem cells, decreased proliferation, and impaired maturation of immature neurons. Klotho-deficient neurospheres show reduced proliferation and size that is rescued by supplementation with shed klotho protein. Conversely, 6-month-old klotho-overexpressing mice exhibit increased numbers of neural stem cells, increased proliferation, and more immature neurons with enhanced dendritic arborization. Protection from normal age-related loss of object location memory with klotho overexpression and loss of spatial memory when klotho is reduced by even half suggests direct, local effects of the protein. Together, these data show that klotho is a novel regulator of postnatal neurogenesis affecting neural stem cell proliferation and maturation sufficient to impact hippocampal-dependent spatial memory function.
Collapse
Affiliation(s)
- Ann M Laszczyk
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephanie Fox-Quick
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hai T Vo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dailey Nettles
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Phyllis C Pugh
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Gwendalyn D King
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
80
|
Abulizi P, Zhou XH, Keyimu K, Luo M, Jin FQ. Correlation between KLOTHO gene and mild cognitive impairment in the Uygur and Han populations of Xinjiang. Oncotarget 2017; 8:75174-75185. [PMID: 29088855 PMCID: PMC5650410 DOI: 10.18632/oncotarget.20655] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 07/18/2017] [Indexed: 01/03/2023] Open
Abstract
The secretive Klotho protein is an anti-aging regulatory hormone that plays a physiological role in many target organs. The present study aims to investigate the correlation between Klotho gene and mild cognitive impairment (MCI) in Uygur and Han populations in Xinjiang. From July 2008 to April 2014, stratified random multistage cluster sampling was used in combination with the methods of on-site questionnaire and household survey to conduct a cross-sectional MCI investigation on selected Uygur and Han subjects aged over 60 years old in Xinjiang region. 323 Uygur and Han MCI patients were randomly selected and matched with 343 subjects in the normal control group. SNaPshot technique was used to detect the polymorphisms of Klotho gene. A case-control associated analysis was conducted to analyze the genotype and allele frequencies of single nucleotide polymorphisms (SNPs) in the MCI group and the normal control group. The polymorphisms of rs1207568 and rs9536314/rs9527025 loci in Klotho gene were different among MCI populations in Xinjiang, and after group assignments based on ethnic background, the polymorphisms of rs1207568 and rs9536314/rs9527025 loci were associated with the Uygur MCI population and were not relevant to the Han MIC population. The frequencies of mutational alleles of rs9536314/rs9527025 locus in the Uygur population were significantly higher than those in the Han population. The genotype and allele frequencies of rs1207568 locus in the Uygur and Han populations were similar. The polymorphisms of rs1207568 and rs9536314/rs9527025 loci in Klotho gene may be associated with the Uygur MCI population in Xinjiang.
Collapse
Affiliation(s)
- Palida Abulizi
- No1 Cadre Wards Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Xiao-Hui Zhou
- No1 Cadre Wards Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Kabinuer Keyimu
- No1 Cadre Wards Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Mei Luo
- No1 Cadre Wards Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Feng-Qing Jin
- No1 Cadre Wards Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| |
Collapse
|
81
|
Boksha IS, Prokhorova TA, Savushkina OK, Tereshkina EB. Klotho protein: Its role in aging and central nervous system pathology. BIOCHEMISTRY (MOSCOW) 2017; 82:990-1005. [DOI: 10.1134/s0006297917090024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
82
|
Karami M, Mehrabi F, Allameh A, Pahlevan Kakhki M, Amiri M, Emami Aleagha MS. Klotho gene expression decreases in peripheral blood mononuclear cells (PBMCs) of patients with relapsing-remitting multiple sclerosis. J Neurol Sci 2017; 381:305-307. [PMID: 28991703 DOI: 10.1016/j.jns.2017.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/21/2017] [Accepted: 09/08/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND we recently showed that a hypothesized anti-aging and anti-inflammatory protein, namely Klotho, may contribute to the etiology and/or pathogenesis of multiple sclerosis (MS). In addition, Klotho function and its gene expression are dependent on inflammatory pathways. Accordingly, the aim of this study was to investigate the Klotho gene expression within peripheral blood mononuclear cells (PBMCs) of patients with MS. METHODS Altogether, 30 patients with relapsing-remitting MS (RRMS) along with 30 age and sex-matched healthy individuals were enrolled in this study. Blood samples were obtained from all participants and then PBMCs were isolated. The quantitative Real-Time PCR was carried out for Klotho mRNA derived from PBMCs. RESULTS The results showed that klotho gene expression in the PBMCs of patients with RRMS is nearly 2.5-fold less than healthy individuals (P=0.0006). CONCLUSION This is the first study demonstrating a possible role of Klotho in the PBMCs of MS patients.
Collapse
Affiliation(s)
- Masoumeh Karami
- Department of Biochemistry, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Farzad Mehrabi
- Department of Neurology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Majid Pahlevan Kakhki
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Amiri
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | | |
Collapse
|
83
|
Klotho, APOEε4, cognitive ability, brain size, atrophy, and survival: a study in the Aberdeen Birth Cohort of 1936. Neurobiol Aging 2017; 55:91-98. [DOI: 10.1016/j.neurobiolaging.2017.02.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 02/24/2017] [Accepted: 02/27/2017] [Indexed: 01/03/2023]
|
84
|
Olauson H, Mencke R, Hillebrands JL, Larsson TE. Tissue expression and source of circulating αKlotho. Bone 2017; 100:19-35. [PMID: 28323144 DOI: 10.1016/j.bone.2017.03.043] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 12/16/2022]
Abstract
αKlotho (Klotho), a type I transmembrane protein and a coreceptor for Fibroblast Growth Factor-23, was initially thought to be expressed only in a limited number of tissues, most importantly the kidney, parathyroid gland and choroid plexus. Emerging data may suggest a more ubiquitous Klotho expression pattern which has prompted reevaluation of the restricted Klotho paradigm. Herein we systematically review the evidence for Klotho expression in various tissues and cell types in humans and other mammals, and discuss potential reasons behind existing conflicting data. Based on current literature and tissue expression atlases, we propose a classification of tissues into high, intermediate and low/absent Klotho expression. The functional relevance of Klotho in organs with low expression levels remain uncertain and there is currently limited data on a role for membrane-bound Klotho outside the kidney. Finally, we review the evidence for the tissue source of soluble Klotho, and conclude that the kidney is likely to be the principal source of circulating Klotho in physiology.
Collapse
Affiliation(s)
- Hannes Olauson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Rik Mencke
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tobias E Larsson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
85
|
The relevance of α-KLOTHO to the central nervous system: Some key questions. Ageing Res Rev 2017; 36:137-148. [PMID: 28323064 DOI: 10.1016/j.arr.2017.03.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/10/2017] [Accepted: 03/16/2017] [Indexed: 12/20/2022]
Abstract
α-Klotho is well described as an anti-aging protein, with critical roles in kidney function as a transmembrane co-receptor for FGF23, and as a soluble factor in serum. α-Klotho is also expressed in the choroid plexus, where it is released into the cerebrospinal fluid. Nonetheless, α-Klotho is also expressed in the brain parenchyma. Accumulating evidence indicates that this pool of α-Klotho, which we define as brain α-Klotho, may play important roles as a neuroprotective factor and in promoting myelination, thereby supporting healthy brain aging. Here we summarize what is known about brain α-Klotho before focusing on the outstanding scientific questions related to its function. We believe there is a need for in vitro studies designed to distinguish between brain α-Klotho and other pools of α-Klotho, and for a greater understanding of the basic function of soluble α-Klotho. The mechanism by which the human KL-VS variant affects cognition also requires further elucidation. To help address these questions we suggest some experimental approaches that other laboratories might consider. In short, we hope to stimulate fresh ideas and encourage new research approaches that will allow the importance of α-Klotho for the aging brain to become clear.
Collapse
|
86
|
Li Q, Vo HT, Wang J, Fox-Quick S, Dobrunz LE, King GD. Klotho regulates CA1 hippocampal synaptic plasticity. Neuroscience 2017; 347:123-133. [PMID: 28215989 PMCID: PMC5392240 DOI: 10.1016/j.neuroscience.2017.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 02/02/2017] [Accepted: 02/05/2017] [Indexed: 01/07/2023]
Abstract
Global klotho overexpression extends lifespan while global klotho-deficiency shortens it. As well, klotho protein manipulations inversely regulate cognitive function. Mice without klotho develop rapid onset cognitive impairment before they are 2months old. Meanwhile, adult mice overexpressing klotho show enhanced cognitive function, particularly in hippocampal-dependent tasks. The cognitive enhancing effects of klotho extend to humans with a klotho polymorphism that increases circulating klotho and executive function. To affect cognitive function, klotho could act in or on the synapse to modulate synaptic transmission or plasticity. However, it is not yet known if klotho is located at synapses, and little is known about its effects on synaptic function. To test this, we fractionated hippocampi and detected klotho expression in both pre and post-synaptic compartments. We find that loss of klotho enhances both pre and post-synaptic measures of CA1 hippocampal synaptic plasticity at 5weeks of age. However, a rapid loss of synaptic enhancement occurs such that by 7weeks, when mice are cognitively impaired, there is no difference from wild-type controls. Klotho overexpressing mice show no early life effects on synaptic plasticity, but decreased CA1 hippocampal long-term potentiation was measured at 6months of age. Together these data suggest that klotho affects cognition, at least in part, by regulating hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Qin Li
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hai T Vo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jing Wang
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Stephanie Fox-Quick
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lynn E Dobrunz
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gwendalyn D King
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
87
|
Kunert SK, Hartmann H, Haffner D, Leifheit-Nestler M. Klotho and fibroblast growth factor 23 in cerebrospinal fluid in children. J Bone Miner Metab 2017; 35:215-226. [PMID: 27017221 DOI: 10.1007/s00774-016-0746-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/15/2016] [Indexed: 12/13/2022]
Abstract
The fibroblast growth factor (FGF) 23/Klotho axis is a principal regulator of phosphate hemostasis and vitamin D metabolism, but limited data is available on its role in the central nervous system. Here, we investigate soluble α-Klotho (sKlotho) and C-terminal as well as intact FGF23 in cerebrospinal fluid (CSF) and plasma and their relationship to mineral metabolism parameters in humans. In 39 children aged 0.3-16.8 years undergoing lumbar puncture for the exclusion of inflammatory neurological disease, sKlotho and FGF23 were investigated by Western blot analysis, followed by ELISA quantification in CSF and plasma. The percentage of intrathecal synthesis of both proteins was calculated by measuring both the expected and observed CSF/plasma ratios of sKlotho and FGF23. The secreted (KL1) and cleaved (KL1+KL2) isoforms of sKlotho, and FGF23 were clearly detected in CSF in all subjects, although protein levels were lower compared to those of plasma samples (each p < 0.01). The intrathecal percentage of CSF sKlotho and FGF23 synthesis amounted to 98 and 99 %, respectively. CSF sKlotho levels were higher in boys than in girls (p < 0.01), and correlated positively with plasma C-terminal FGF23 concentrations (p < 0.05) and standardized height (p < 0.01). Importantly, there were no significant correlations between plasma and CSF levels of sKlotho or FGF23. Plasma sKlotho as well as C-terminal and intact FGF23, respectively, were associated with parameters of mineral metabolism These results provide evidence that cleaved and secreted sKlotho and FGF23 are present in CSF, mainly derived from brain and affected by sex, height, and mineral metabolism parameters in children. Nevertheless, the absence of significant associations between plasma and CSF levels of Klotho and FGF23, respectively, suggest that the regulation of Klotho and FGF23 may be different between organs secreting these hormones into blood and CSF.
Collapse
Affiliation(s)
- Svenja Kristin Kunert
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Hans Hartmann
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Maren Leifheit-Nestler
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
88
|
Zhang Y, Wang L, Wu Z, Yu X, Du X, Li X. The Expressions of Klotho Family Genes in Human Ocular Tissues and in Anterior Lens Capsules of Age-Related Cataract. Curr Eye Res 2017; 42:871-875. [PMID: 28095050 DOI: 10.1080/02713683.2016.1259421] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Yu Zhang
- Department of Laboratory, Henan Eye Institute, Henan Eye Hospital, Zhengzhou, China
| | - Liya Wang
- Department of Keratopathy, Henan Eye Institute, Henan Eye Hospital, Zhengzhou, China
| | - Zhong Wu
- Department of Cataract, Henan Eye Institute, Henan Eye Hospital, Zhengzhou, China
| | - Xiaolin Yu
- Department of Cataract, Henan Eye Institute, Henan Eye Hospital, Zhengzhou, China
| | - Xiaofeng Du
- Eye Bank, Henan Eye Institute, Henan Eye Hospital, Zhengzhou, China
| | - Xiaohua Li
- Department of Laboratory, Henan Eye Institute, Henan Eye Hospital, Zhengzhou, China
| |
Collapse
|
89
|
Cheng X, Xie B, Qi J, Zhao X, Zhang Z, Qiu M, Yang J. Rat astrocytes are more supportive for mouse OPC self-renewal than mouse astrocytes in culture. Dev Neurobiol 2016; 77:907-916. [PMID: 28033654 DOI: 10.1002/dneu.22476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/27/2016] [Accepted: 12/09/2016] [Indexed: 11/08/2022]
Abstract
Mouse primary oligodendrocyte precursor cells (OPCs) are increasingly used to study the molecular mechanisms underlying the phenotype changes in oligodendrocyte differentiation and axonal myelination observed in transgenic or mutant mouse models. However, mouse OPCs are much more difficult to be isolated by the simple dissociation culture of brain tissues than their rat counterparts. To date, the mechanisms underlying the species difference in OPC preparation remain obscure. In this study, we showed that astrocytes from rats have a stronger effect than those from mouse in promoting OPC proliferation and survival in vitro. Mouse astrocytes displayed significantly weaker viability in culture and reduced potential in maintaining OPC self-renewal, as confirmed by culturing OPCs with conditioned media from rat or mouse astrocytes. These results explained the reason for why stratified cultures of OPCs and astrocytes are difficult to be achieved in mouse CNS tissues. Based on these findings, we adopted inactivated rat astrocytes as feeder cells to support the self-renewal of mouse cortical OPCs and preparation of high-purity mouse OPCs. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 907-916, 2017.
Collapse
Affiliation(s)
- Xuejun Cheng
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environment Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Binghua Xie
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environment Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Jiajun Qi
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environment Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Xiaofeng Zhao
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environment Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Zunyi Zhang
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environment Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Mengsheng Qiu
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environment Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China.,Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky, 40292
| | - Junlin Yang
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environment Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| |
Collapse
|
90
|
Abe H, Saito F, Tanaka T, Mizukami S, Watanabe Y, Imatanaka N, Akahori Y, Yoshida T, Shibutani M. Global gene expression profiles in brain regions reflecting abnormal neuronal and glial functions targeting myelin sheaths after 28-day exposure to cuprizone in rats. Toxicol Appl Pharmacol 2016; 310:20-31. [DOI: 10.1016/j.taap.2016.08.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 07/28/2016] [Accepted: 08/09/2016] [Indexed: 10/21/2022]
|
91
|
Mirza SB, Ekhteiari Salmas R, Fatmi MQ, Durdagi S. Discovery of Klotho peptide antagonists against Wnt3 and Wnt3a target proteins using combination of protein engineering, protein-protein docking, peptide docking and molecular dynamics simulations. J Enzyme Inhib Med Chem 2016; 32:84-98. [PMID: 27766889 PMCID: PMC6009926 DOI: 10.1080/14756366.2016.1235569] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The Klotho is known as lifespan enhancing protein involved in antagonizing the effect of Wnt proteins. Wnt proteins are stem cell regulators, and uninterrupted exposure of Wnt proteins to the cell can cause stem and progenitor cell senescence, which may lead to aging. Keeping in mind the importance of Klotho in Wnt signaling, in silico approaches have been applied to study the important interactions between Klotho and Wnt3 and Wnt3a (wingless-type mouse mammary tumor virus (MMTV) integration site family members 3 and 3a). The main aim of the study is to identify important residues of the Klotho that help in designing peptides which can act as Wnt antagonists. For this aim, a protein engineering study is performed for Klotho, Wnt3 and Wnt3a. During the theoretical analysis of homology models, unexpected role of number of disulfide bonds and secondary structure elements has been witnessed in case of Wnt3 and Wnt3a proteins. Different in silico experiments were carried out to observe the effect of correct number of disulfide bonds on 3D protein models. For this aim, total of 10 molecular dynamics (MD) simulations were carried out for each system. Based on the protein–protein docking simulations of selected protein models of Klotho with Wnt3 and Wnt3a, different peptides derived from Klotho have been designed. Wnt3 and Wnt3a proteins have three important domains: Index finger, N-terminal domain and a patch of ∼10 residues on the solvent exposed surface of palm domain. Protein–peptide docking of designed peptides of Klotho against three important domains of palmitoylated Wnt3 and Wnt3a yields encouraging results and leads better understanding of the Wnt protein inhibition by proposed Klotho peptides. Further in vitro studies can be carried out to verify effects of novel designed peptides as Wnt antagonists.
Collapse
Affiliation(s)
- Shaher Bano Mirza
- a Department of Biophysics, School of Medicine , Bahcesehir University (BAU) , Istanbul , Turkey.,b Department of Biosciences , COMSATS Institute of Information Technology (CIIT) , Islamabad , Pakistan
| | - Ramin Ekhteiari Salmas
- a Department of Biophysics, School of Medicine , Bahcesehir University (BAU) , Istanbul , Turkey
| | - M Qaiser Fatmi
- b Department of Biosciences , COMSATS Institute of Information Technology (CIIT) , Islamabad , Pakistan
| | - Serdar Durdagi
- a Department of Biophysics, School of Medicine , Bahcesehir University (BAU) , Istanbul , Turkey
| |
Collapse
|
92
|
Garg M, Chawla M, Chunduri V, Kumar R, Sharma S, Sharma NK, Kaur N, Kumar A, Mundey JK, Saini MK, Singh SP. Transfer of grain colors to elite wheat cultivars and their characterization. J Cereal Sci 2016. [DOI: 10.1016/j.jcs.2016.08.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
93
|
Yang J, Cheng X, Shen J, Xie B, Zhao X, Zhang Z, Cao Q, Shen Y, Qiu M. A Novel Approach for Amplification and Purification of Mouse Oligodendrocyte Progenitor Cells. Front Cell Neurosci 2016; 10:203. [PMID: 27597818 PMCID: PMC4992724 DOI: 10.3389/fncel.2016.00203] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022] Open
Abstract
Although transgenic and knockout mice are widely used to study the specification and differentiation of oligodendrocyte precursor cells (OPCs), mouse primary OPCs are difficult to be purified and maintained, and many in vitro studies have to resort to rat OPCs as substitutes. In this study, we reported that mouse O4 negative early-stage OPCs can be obtained by culturing cortical tissue blocks, and the simultaneous treatment of OPCs with Platelet Derived Growth Factor-AA (PDGFaa), basic fibroblast growth factor (bFGF), and epidermal growth factor (EGF) is the key for the propagation of mouse OPCs in culture. EGF was found to be a potent mitogen for OPCs and cooperate with PDGFaa to extend cell division and inhibit their differentiation. EGF also collaborates with PDGFaa and bFGF to convert bipolar or tripolar OPCs to more vital fibroblast-like OPCs without compromising their oligodendrocyte differentiation potential. In addition, EGF promoted the survival and proliferation of glial progenitor cells (GPCs) derived from primary OPC cultures, and a mixture of GPCs and OPCs can be obtained and propagated in the presence of EGF, bFGF, and PDGFaa. Once EGF is withdrawn, GPC population decreased sharply and fibroblast-like OPCs changed into typical OPCs morphology, then homogeneous OPCs were obtained subsequently.
Collapse
Affiliation(s)
- Junlin Yang
- Zhejiang Key Laboratory of Organ Development and Regeneration, The Institute of Developmental and Regenerative Biology, College of Life and Environment Sciences, Hangzhou Normal University Hangzhou, China
| | - Xuejun Cheng
- Zhejiang Key Laboratory of Organ Development and Regeneration, The Institute of Developmental and Regenerative Biology, College of Life and Environment Sciences, Hangzhou Normal University Hangzhou, China
| | - Jiaxi Shen
- Zhejiang Key Laboratory of Organ Development and Regeneration, The Institute of Developmental and Regenerative Biology, College of Life and Environment Sciences, Hangzhou Normal University Hangzhou, China
| | - Binghua Xie
- Zhejiang Key Laboratory of Organ Development and Regeneration, The Institute of Developmental and Regenerative Biology, College of Life and Environment Sciences, Hangzhou Normal University Hangzhou, China
| | - Xiaofeng Zhao
- Zhejiang Key Laboratory of Organ Development and Regeneration, The Institute of Developmental and Regenerative Biology, College of Life and Environment Sciences, Hangzhou Normal University Hangzhou, China
| | - Zunyi Zhang
- Zhejiang Key Laboratory of Organ Development and Regeneration, The Institute of Developmental and Regenerative Biology, College of Life and Environment Sciences, Hangzhou Normal University Hangzhou, China
| | - Qilin Cao
- The Vivian L Smith Department of Neurosurgery, University of Texas Medical School at Houston, Houston TX, USA
| | - Ying Shen
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine Hangzhou, China
| | - Mengsheng Qiu
- Zhejiang Key Laboratory of Organ Development and Regeneration, The Institute of Developmental and Regenerative Biology, College of Life and Environment Sciences, Hangzhou Normal UniversityHangzhou, China; Department of Anatomical Sciences and Neurobiology, University of Louisville, LouisvilleKY, USA
| |
Collapse
|
94
|
Bahrami SA, Bakhtiari N. Ursolic acid regulates aging process through enhancing of metabolic sensor proteins level. Biomed Pharmacother 2016; 82:8-14. [PMID: 27470332 DOI: 10.1016/j.biopha.2016.04.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 04/23/2016] [Accepted: 04/25/2016] [Indexed: 02/06/2023] Open
Abstract
We previously reported that Ursolic Acid (UA) ameliorates skeletal muscle performance through satellite cells proliferation and cellular energy status. In studying the potential role of the hypothalamus in aging, we developed a strategy to pursue UA effects on the hypothalamus anti-aging proteins such as; SIRT1, SIRT6, PGC-1β and α-Klotho. In this study, we used a model of aging animals (C57BL/6). UA dissolved in Corn oil (20mg/ml) and then administrated (200mg/Kg i.p injection) to mice, twice daily for 7days. After treatment times, the mice perfused and the hypothalamus isolated for preparing of tissue to Immunofluorescence microscopy. The data illustrated that UA significantly increased SIRT1 (∼3.5±0.3 folds) and SIRT-6 (∼1.5±0.2 folds) proteins overexpression (P<0.001). In addition, our results showed that UA enhanced α-Klotho (∼3.3±0.3) and PGC-1β (∼2.6±0.2 folds) proteins levels (P<0. 01). In this study, data were analyzed using SPSS 16 (ANOVA test). To the best of our knowledge, it seems that UA through enhancing of anti-aging biomarkers (SIRT1 and SIRT6) and PGC-1β in hypothalamus regulates aging-process and attenuates mitochondrial-related diseases. In regard to the key role of α-Klotho in aging, our data indicate that UA may be on the horizon to forestall diseases of aging.
Collapse
Affiliation(s)
- Soroush Alaghehband Bahrami
- Department of Biochemistry, Faculty of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Nuredin Bakhtiari
- Department of Biochemistry, Faculty of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran.
| |
Collapse
|
95
|
Sárvári M, Kalló I, Hrabovszky E, Solymosi N, Rodolosse A, Liposits Z. Long-Term Estrogen Receptor Beta Agonist Treatment Modifies the Hippocampal Transcriptome in Middle-Aged Ovariectomized Rats. Front Cell Neurosci 2016; 10:149. [PMID: 27375434 PMCID: PMC4901073 DOI: 10.3389/fncel.2016.00149] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/27/2016] [Indexed: 11/13/2022] Open
Abstract
Estradiol (E2) robustly activates transcription of a broad array of genes in the hippocampal formation of middle-aged ovariectomized rats via estrogen receptors (ERα, ERβ, and G protein-coupled ER). Selective ERβ agonists also influence hippocampal functions, although their downstream molecular targets and mechanisms are not known. In this study, we explored the effects of long-term treatment with ERβ agonist diarylpropionitrile (DPN, 0.05 mg/kg/day, sc.) on the hippocampal transcriptome in ovariectomized, middle-aged (13 month) rats. Isolated hippocampal formations were analyzed by Affymetrix oligonucleotide microarray and quantitative real-time PCR. Four hundred ninety-seven genes fulfilled the absolute fold change higher than 2 (FC > 2) selection criterion. Among them 370 genes were activated. Pathway analysis identified terms including glutamatergic and cholinergic synapse, RNA transport, endocytosis, thyroid hormone signaling, RNA degradation, retrograde endocannabinoid signaling, and mRNA surveillance. PCR studies showed transcriptional regulation of 58 genes encoding growth factors (Igf2, Igfb2, Igf1r, Fgf1, Mdk, Ntf3, Bdnf), transcription factors (Otx2, Msx1), potassium channels (Kcne2), neuropeptides (Cck, Pdyn), peptide receptors (Crhr2, Oprm1, Gnrhr, Galr2, Sstr1, Sstr3), neurotransmitter receptors (Htr1a, Htr2c, Htr2a, Gria2, Gria3, Grm5, Gabra1, Chrm5, Adrb1), and vesicular neurotransmitter transporters (Slc32a1, Slc17a7). Protein-protein interaction analysis revealed networking of clusters associated with the regulation of growth/troph factor signaling, transcription, translation, neurotransmitter and neurohormone signaling mechanisms and potassium channels. Collectively, the results reveal the contribution of ERβ-mediated processes to the regulation of transcription, translation, neurogenesis, neuromodulation, and neuroprotection in the hippocampal formation of ovariectomized, middle-aged rats and elucidate regulatory channels responsible for DPN-altered functional patterns. These findings support the notion that selective activation of ERβ may be a viable approach for treating the neural symptoms of E2 deficiency in menopause.
Collapse
Affiliation(s)
- Miklós Sárvári
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences Budapest, Hungary
| | - Imre Kalló
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of SciencesBudapest, Hungary; Faculty of Information Technology and Bionics, Pázmány Péter Catholic UniversityBudapest, Hungary
| | - Erik Hrabovszky
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences Budapest, Hungary
| | - Norbert Solymosi
- Faculty of Veterinary Science, Szent István University Budapest, Hungary
| | - Annie Rodolosse
- Functional Genomics Core, Institute for Research in Biomedicine Barcelona, Spain
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of SciencesBudapest, Hungary; Faculty of Information Technology and Bionics, Pázmány Péter Catholic UniversityBudapest, Hungary
| |
Collapse
|
96
|
Leibrock CB, Voelkl J, Kuro-O M, Lang F, Lang UE. 1,25(OH)2D3 dependent overt hyperactivity phenotype in klotho-hypomorphic mice. Sci Rep 2016; 6:24879. [PMID: 27109615 PMCID: PMC4843009 DOI: 10.1038/srep24879] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/05/2016] [Indexed: 02/06/2023] Open
Abstract
Klotho, a protein mainly expressed in kidney and cerebral choroid plexus, is a powerful regulator of 1,25(OH)2D3 formation. Klotho-deficient mice (kl/kl) suffer from excessive plasma 1,25(OH)2D3-, Ca2+- and phosphate-concentrations, leading to severe soft tissue calcification and accelerated aging. NH4Cl treatment prevents tissue calcification and premature ageing without affecting 1,25(OH)2D3-formation. The present study explored the impact of excessive 1,25(OH)2D3 formation in NH4Cl-treated kl/kl-mice on behavior. To this end kl/kl-mice and wild-type mice were treated with NH4Cl and either control diet or vitamin D deficient diet (LVD). As a result, plasma 1,25(OH)2D3-, Ca2+- and phosphate-concentrations were significantly higher in untreated and in NH4Cl-treated kl/kl-mice than in wild-type mice, a difference abrogated by LVD. In each, open field, dark-light box, and O-maze NH4Cl-treated kl/kl-mice showed significantly higher exploratory behavior than untreated wild-type mice, a difference abrogated by LVD. The time of floating in the forced swimming test was significantly shorter in NH4Cl treated kl/kl-mice compared to untreated wild-type mice and to kl/kl-mice on LVD. In wild-type animals, NH4Cl treatment did not significantly alter 1,25(OH)2D3, calcium and phosphate concentrations or exploratory behavior. In conclusion, the excessive 1,25(OH)2D3 formation in klotho-hypomorphic mice has a profound effect on murine behavior.
Collapse
Affiliation(s)
- Christina B Leibrock
- Department of Physiology, Cardiology &Vascular Medicine, University of Tübingen, Gmelinstr. 5, 72076 Tübingen, Germany
| | - Jakob Voelkl
- Department of Physiology, Cardiology &Vascular Medicine, University of Tübingen, Gmelinstr. 5, 72076 Tübingen, Germany
| | - Makoto Kuro-O
- Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Florian Lang
- Department of Physiology, Cardiology &Vascular Medicine, University of Tübingen, Gmelinstr. 5, 72076 Tübingen, Germany
| | - Undine E Lang
- Department of Psychiatry, University of Basel, Wilhelm Klein-Strasse 27, CH-4012 Basel, Switzerland
| |
Collapse
|
97
|
Nam KN, Mounier A, Fitz NF, Wolfe C, Schug J, Lefterov I, Koldamova R. RXR controlled regulatory networks identified in mouse brain counteract deleterious effects of Aβ oligomers. Sci Rep 2016; 6:24048. [PMID: 27051978 PMCID: PMC4823697 DOI: 10.1038/srep24048] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/16/2016] [Indexed: 11/21/2022] Open
Abstract
Bexarotene, a selective agonist for Retinoid X receptors (RXR) improves cognitive deficits and amyloid-β (Aβ) clearance in mice. Here we examine if the effect of bexarotene on RXR cistrome and transcriptomes depend on APOE isoform and Aβ deposition. We found bexarotene increased RXR binding to promoter regions in cortex of APOE3 mice. Rho family GTPases and Wnt signaling pathway were highly enriched in ChIP-seq and RNA-seq datasets and members of those pathways - Lrp1, Lrp5, Sfrp5 and Sema3f were validated. The effect of APOE isoform was compared in APOE3 and APOE4 mice and we found significant overlapping in affected pathways. ChIP-seq using mouse embryonic stem cells and enrichment levels of histone marks H3K4me3 and H3K27me3 revealed that, bexarotene induced epigenetic changes, consistent with increased neuronal differentiation and in correlation with changes in transcription. Comparison of transcriptome in APOE3 and APP/APOE3 mice revealed that amyloid deposition significantly affects the response to bexarotene. In primary neurons, bexarotene ameliorated the damaged dendrite complexity and loss of neurites caused by Aβ42. Finally, we show that the disruption of actin cytoskeleton induced by Aβ42in vitro was inhibited by bexarotene treatment. Our results suggest a mechanism to establish RXR therapeutic targets with significance in neurodegeneration.
Collapse
Affiliation(s)
- Kyong Nyon Nam
- Department of Environmental &Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Anais Mounier
- Department of Environmental &Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Nicholas F Fitz
- Department of Environmental &Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Cody Wolfe
- Department of Environmental &Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Jonathan Schug
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Iliya Lefterov
- Department of Environmental &Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Radosveta Koldamova
- Department of Environmental &Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| |
Collapse
|
98
|
Abraham CR, Mullen PC, Tucker-Zhou T, Chen CD, Zeldich E. Klotho Is a Neuroprotective and Cognition-Enhancing Protein. VITAMINS AND HORMONES 2016; 101:215-38. [PMID: 27125744 DOI: 10.1016/bs.vh.2016.02.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this chapter, we will describe what has been learned about Klotho and its potential functions in the brain. Klotho is localized in the choroid plexus and, to a lesser extent, in hippocampal neurons. Cognitive decline is a common issue in human aging affecting over 50% of the population. This cognitive decline can also be seen in animal models such as the Rhesus monkey. A long-term study undertaken by our lab demonstrated that normal brain aging in rhesus monkeys and other animal models is associated with a significant downregulation of Klotho expression. This observation substantiates data from other laboratories that have reported that loss of Klotho accelerates the development of aging-like phenotypes, including cognitive deficits, whereas Klotho overexpression extends life span and enhances cognition in mice and humans. Klotho is a type 1 transmembrane pleiotropic protein predominantly expressed in kidney and brain and shed by ADAM 10 and 17 into the blood and cerebral spinal fluid, respectively. While the renal functions of Klotho are well known, its roles in the brain remain to be fully elucidated. We recently demonstrated that Klotho protects hippocampal neurons from amyloid and glutamate toxicity via the activation of an antioxidant enzymatic system suggesting Klotho is a neuroprotective protein. Furthermore, Klotho is necessary for oligodendrocyte maturation and myelin integrity. Through its diverse roles in the brain, Klotho has become a new therapeutic target for neurodegenerative diseases such as Alzheimer's disease and demyelinating diseases like multiple sclerosis. Discovery of small molecule Klotho enhancers may lead to novel treatments for these incurable disorders.
Collapse
Affiliation(s)
- C R Abraham
- Boston University School of Medicine, Boston, MA, United States.
| | - P C Mullen
- Boston University School of Medicine, Boston, MA, United States
| | - T Tucker-Zhou
- Boston University School of Medicine, Boston, MA, United States
| | - C D Chen
- Boston University School of Medicine, Boston, MA, United States
| | - E Zeldich
- Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
99
|
Abstract
There is increasing evidence that a deficiency in vitamin D contributes to many human diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), hypertension and cardiovascular disease. The ability of vitamin D to maintain healthy cells seems to depend on its role as a guardian of phenotypic stability particularly with regard to the reactive oxygen species (ROS) and Ca2+ signalling systems. Vitamin D maintains the expression of those signalling components responsible for stabilizing the low-resting state of these two signalling pathways. This vitamin D signalling stability hypothesis proposes that vitamin D, working in conjunction with klotho and Nrf2 (nuclear factor-erythroid-2-related factor 2), acts as a custodian to maintain the normal function of the ROS and Ca2+ signalling pathways. A decline in vitamin D levels will lead to an erosion of this signalling stability and may account for why so many of the major diseases in man, which have been linked to vitamin D deficiency, are associated with a dysregulation in both ROS and Ca2+ signalling.
Collapse
|
100
|
Ahmadi M, Emami Aleagha MS, Harirchian MH, Yarani R, Tavakoli F, Siroos B. Multiple sclerosis influences on the augmentation of serum Klotho concentration. J Neurol Sci 2016; 362:69-72. [PMID: 26944121 DOI: 10.1016/j.jns.2016.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/04/2016] [Accepted: 01/05/2016] [Indexed: 11/17/2022]
Abstract
We have already shown that the concentration of secreted form of Klotho decreases in the cerebrospinal fluid of patients with relapsing-remitting multiple sclerosis (RRMS). The current study aimed at assessing possible changes in the serum Klotho concentration of MS patients. Participants involved 15 new cases of RRMS patients in the relapse phase, 15 RRMS patients who had been suffering from the disease for more than three years and were under regular treatments (interferon beta-1a) and, finally, 15 non-MS patients who constituted the control group. Beside thorough neurological examinations, demographic and clinical data (e.g. gender, age, duration of disease and expanded disability status scale) were obtained. Serum Klotho concentration was measured using ELISA method. The results showed no statistically meaningful difference between new cases of RRMS (585.56pg/ml±153.99) and control group (556.81pg/ml±120.36; P=0.859). The serum Klotho level, however, was significantly higher in patients with prolonged disease duration (696.94pg/ml±170.52; P=0.037) in comparison with the subjects in the control group. In conclusion, this study showed that serum Klotho concentration tends to be higher in MS patients when compared to control group. This finding might be attributed to treatment of MS patients with immunomodulatory drugs or a compensatory response to enhance CNS regeneration and/or vitamin D biosynthesis. Further studies are required to elucidate the role of Klotho in MS pathophysiology.
Collapse
Affiliation(s)
- Mona Ahmadi
- Iranian Centre of Neurological Research, Department of Neurology Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sajad Emami Aleagha
- Iranian Centre of Neurological Research, Department of Neurology Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran; Department of Clinical Biochemistry, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Hossein Harirchian
- Iranian Centre of Neurological Research, Department of Neurology Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Yarani
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| | - Farhad Tavakoli
- Department of Anesthesiology, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahaadin Siroos
- Iranian Centre of Neurological Research, Department of Neurology Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|