151
|
Vasilopoulou MΑ, Ioannou E, Roussis V, Chondrogianni N. Modulation of the ubiquitin-proteasome system by marine natural products. Redox Biol 2021; 41:101897. [PMID: 33640701 PMCID: PMC7921624 DOI: 10.1016/j.redox.2021.101897] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
The ubiquitin-proteasome system (UPS) is a key player in the maintenance of cellular protein homeostasis (proteostasis). Since proteasome function declines upon aging leading to the acceleration of its progression and the manifestation of age-related pathologies, many attempts have been performed towards proteasome activation as a strategy to promote healthspan and longevity. The marine environment hosts a plethora of organisms that produce a vast array of primary and secondary metabolites, the majority of which are unique, exhibiting a wide spectrum of biological activities. The fact that these biologically important compounds are also present in edible marine organisms has sparked the interest for elucidating their potential health-related applications. In this review, we focus on the antioxidant, anti-aging, anti-aggregation and anti-photoaging properties of various marine constituents. We further discuss representatives of marine compounds classes with regard to their potential (direct or indirect) action on UPS components that could serve as UPS modulators and exert beneficial effects on conditions such as oxidative stress, aging and age-related diseases.
Collapse
Affiliation(s)
- Mary Α Vasilopoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., Athens, 11635, Greece; Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500, Larisa, Greece.
| | - Efstathia Ioannou
- Section of Pharmacognosy and Chemistry of Natural Products, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, Athens, 15771, Greece.
| | - Vassilios Roussis
- Section of Pharmacognosy and Chemistry of Natural Products, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, Athens, 15771, Greece.
| | - Niki Chondrogianni
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., Athens, 11635, Greece.
| |
Collapse
|
152
|
Sokolov AS, Nekrasov PV, Shaposhnikov MV, Moskalev AA. Hydrogen sulfide in longevity and pathologies: Inconsistency is malodorous. Ageing Res Rev 2021; 67:101262. [PMID: 33516916 DOI: 10.1016/j.arr.2021.101262] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 02/08/2023]
Abstract
Hydrogen sulfide (H2S) is one of the biologically active gases (gasotransmitters), which plays an important role in various physiological processes and aging. Its production in the course of methionine and cysteine catabolism and its degradation are finely balanced, and impairment of H2S homeostasis is associated with various pathologies. Despite the strong geroprotective action of exogenous H2S in C. elegans, there are controversial effects of hydrogen sulfide and its donors on longevity in other models, as well as on stress resistance, age-related pathologies and aging processes, including regulation of senescence-associated secretory phenotype (SASP) and senescent cell anti-apoptotic pathways (SCAPs). Here we discuss that the translation potential of H2S as a geroprotective compound is influenced by a multiplicity of its molecular targets, pleiotropic biological effects, and the overlapping ranges of toxic and beneficial doses. We also consider the challenges of the targeted delivery of H2S at the required dose. Along with this, the complexity of determining the natural levels of H2S in animal and human organs and their ambiguous correlations with longevity are reviewed.
Collapse
|
153
|
Seto M, Weiner RL, Dumitrescu L, Hohman TJ. Protective genes and pathways in Alzheimer's disease: moving towards precision interventions. Mol Neurodegener 2021; 16:29. [PMID: 33926499 PMCID: PMC8086309 DOI: 10.1186/s13024-021-00452-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/20/2021] [Indexed: 12/29/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive, neurodegenerative disorder that is characterized by neurodegeneration, cognitive impairment, and an eventual inability to perform daily tasks. The etiology of Alzheimer's is complex, with numerous environmental and genetic factors contributing to the disease. Late-onset AD is highly heritable (60 to 80%), and over 40 risk loci for AD have been identified via large genome-wide association studies, most of which are common variants with small effect sizes. Although these discoveries have provided novel insight on biological contributors to AD, disease-modifying treatments remain elusive. Recently, the concepts of resistance to pathology and resilience against the downstream consequences of pathology have been of particular interest in the Alzheimer's field as studies continue to identify individuals who evade the pathology of the disease even into late life and individuals who have all of the neuropathological features of AD but evade downstream neurodegeneration and cognitive impairment. It has been hypothesized that a shift in focus from Alzheimer's risk to resilience presents an opportunity to uncover novel biological mechanisms of AD and to identify promising therapeutic targets for the disease. This review will highlight a selection of genes and variants that have been reported to confer protection from AD within the literature and will also discuss evidence for the biological underpinnings behind their protective effect with a focus on genes involved in lipid metabolism, cellular trafficking, endosomal and lysosomal function, synaptic function, and inflammation. Finally, we offer some recommendations in areas where the field can rapidly advance towards precision interventions that leverage the ideas of protection and resilience for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Mabel Seto
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, 1207 17th Ave S, Nashville, TN 37212 USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN USA
| | - Rebecca L. Weiner
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, 1207 17th Ave S, Nashville, TN 37212 USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN USA
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, 1207 17th Ave S, Nashville, TN 37212 USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, 1207 17th Ave S, Nashville, TN 37212 USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN USA
| |
Collapse
|
154
|
Sugar sweetened beverage consumption is positively associated with Klotho levels at two years of age in LatinX youth. BMC Nutr 2021; 7:22. [PMID: 33888157 PMCID: PMC8063295 DOI: 10.1186/s40795-021-00423-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 03/22/2021] [Indexed: 11/21/2022] Open
Abstract
Background Klotho is an anti-aging protein mainly expressed in the kidneys with a smaller amount expressed in adipose tissue. Klotho effects include roles in reducing oxidative stress, insulin signaling, adipogenesis and glucose metabolism. Few studies have investigated the role of dietary factors such as sugar sweetened beverages (SSBs) on serum α-klotho levels in young children. Methods Data was collected from 60 low-income Latina pregnant women and their infants in San Francisco from birth until 2 years of life and examined for associations between dietary factors and child secreted α-klotho protein levels at 2 years. Results Mean α-klotho levels were 1782.96 ± 874.56 pg/mL at 2 years of age. Any consumption of SSBs was independently associated with increased α-klotho levels (Beta = 682.79, 95%CI 67.50, 1298.09; p = 0.03). Household income ranging from $25,000 to $50,000 was also correlated to higher levels of α-klotho in children compared with lower income levels (<$25,000) (Beta = 1613.35, 95%CI 527.37, 2699.33; p = 0.005). Conclusions The positive association between SSB intake and α-klotho levels at 2 years may reflect higher phosphate levels consistent with SSB intake. Higher socioeconomic status may be a proxy for reduced stress exposure in children, also associated with higher α-klotho levels. Future studies should evaluate the early impact of exposures to SSBs, stress and accelerated aging in children.
Collapse
|
155
|
Protective effects of klotho on palmitate-induced podocyte injury in diabetic nephropathy. PLoS One 2021; 16:e0250666. [PMID: 33891667 PMCID: PMC8064606 DOI: 10.1371/journal.pone.0250666] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 04/08/2021] [Indexed: 11/19/2022] Open
Abstract
The anti-aging gene, klotho, has been identified as a multi-functional humoral factor and is implicated in multiple biological processes. However, the effects of klotho on podocyte injury in diabetic nephropathy are poorly understood. Thus, the current study aims to investigate the renoprotective effects of klotho against podocyte injury in diabetic nephropathy. We examined lipid accumulation and klotho expression in the kidneys of diabetic patients and animals. We stimulated cultured mouse podocytes with palmitate to induce lipotoxicity-mediated podocyte injury with or without recombinant klotho. Klotho level was decreased in podocytes of lipid-accumulated obese diabetic kidneys and palmitate-treated mouse podocytes. Palmitate-treated podocytes showed increased apoptosis, intracellular ROS, ER stress, inflammation, and fibrosis, and these were significantly attenuated by klotho administration. Klotho treatment restored palmitate-induced downregulation of the antioxidant molecules, Nrf2, Keap1, and SOD1. Klotho inhibited the phosphorylation of FOXO3a, promoted its nuclear translocation, and then upregulated MnSOD expression. In addition, klotho administration attenuated palmitate-induced cytoskeleton changes, decreased nephrin expression, and increased TRPC6 expression, eventually improving podocyte albumin permeability. These results suggest that klotho administration prevents palmitate-induced functional and morphological podocyte injuries, and this may indicate that klotho is a potential therapeutic agent for the treatment of podocyte injury in obese diabetic nephropathy.
Collapse
|
156
|
Mohanty SK, Suchiang K. Triiodothyronine (T3) enhances lifespan and protects against oxidative stress via activation of Klotho in Caenorhabditis elegans. Biogerontology 2021; 22:397-413. [PMID: 33851304 DOI: 10.1007/s10522-021-09923-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/07/2021] [Indexed: 10/21/2022]
Abstract
Age predisposes individuals to significant diseases, and the biological processes contributing to aging are currently under intense investigation. Klotho is an anti-aging protein with multifaceted roles and is an essential component of the endocrine fibroblast growth factor. In Caenorhabditis elegans (C. elegans), there are two prospective orthologs of α-Klotho, C50F7.10, and E02H9.5, identified. The two orthologs' products are homologous to the highly conserved KL1 domain of human and mouse Klotho protein. Considering the endocrine system's major involvement in an organism's homeostasis and that thyroid disorders increase with advancing age, the molecular mechanisms underlying its impact on different endocrine components during the aging process remain poorly characterized. In this study, we sought to determine the regulatory role of Triiodothyronine (T3) on homologs genes of klotho and its impact on different parameters of aging in the C. elegans model organism. We showed that T3 could increase the mRNA expressions of the klotho homologous genes in C. elegans. Moreover, T3 could also extend a worm lifespan and modulate oxidative stress resistance and aging biomarkers significantly and positively. Further investigations employing different mutant and transgenic strains reveal that these observed effects are mediated through the EGL-17/EGL-15 pathway via Klotho activation along with the involvement of transcription factor DAF-16. In conclusion, these findings have revealed an unexpected link between T3 and Klotho and how this link can modulate the aging process in C. elegans via activation of klotho. This study will help understand the crosstalk and regulations of different endocrine components and their consequences on the aging process in multiple species.
Collapse
Affiliation(s)
- Saswat Kumar Mohanty
- Department of Biochemistry and Molecular Biology, Pondicherry University, Pondicherry, 605 014, India
| | - Kitlangki Suchiang
- Department of Biochemistry and Molecular Biology, Pondicherry University, Pondicherry, 605 014, India.
| |
Collapse
|
157
|
Helvaci N, Kabacam S, Dagdelen S, Lay I, Karabulut E, Mut M, Alikasifoglu M, Erbas T. Klotho gene G395A and C1818T polymorphisms in acromegaly: Association with clinical presentation and comorbidities. Clin Endocrinol (Oxf) 2021; 94:598-605. [PMID: 33296101 DOI: 10.1111/cen.14380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/26/2020] [Accepted: 11/19/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Klotho is a new identified anti-ageing gene with tumour suppressor activities. Current data suggest that there is a tight relationship between Klotho protein and growth hormone (GH)/insulin-like growth factor-1 (IGF-1) axis. PURPOSE This study aimed to investigate the possible association of Klotho gene polymorphisms with acromegaly and to assess whether these polymorphisms contribute to clinical characteristics, comorbidities and biochemical variables in these patients. METHODS The study included 52 patients with acromegaly and 52 unrelated healthy subjects. The Klotho G395A and C1818T polymorphisms were assessed by Sanger sequencing. Serum levels of sKlotho were determined by ELISA method. RESULTS Subjects carrying GA genotype of Klotho G395A polymorphism had 3.27 times higher risk of developing acromegaly [odds ratio (OR), 3.27; 95% confidence interval (CI): 1.37-7.81; p = .023]. The A allele of G395A was significantly associated with acromegaly risk (OR, 2.27; 95% CI: 1.1-4.72; p = .022). No association was observed between the studied polymorphisms and disease characteristics including age at acromegaly diagnosis, size of adenoma, baseline GH and IGF-1 concentrations, and final outcome. G395A polymorphism was associated with the presence of malignancy (OR, 2.24, 95% CI: 1.63-3.08; p = .019) and colorectal polyps (OR, 1.99; 95% CI: 1.02-3.88; p = .047) in patients with acromegaly. Serum sKlotho levels were significantly higher and correlated with GH and IGF-1 levels among acromegaly patients. There was no association between the studied polymorphisms and sKlotho levels. CONCLUSIONS Klotho G395A polymorphism is associated with acromegaly susceptibility and increased risk of malignancy and colorectal polyps in these patients.
Collapse
Affiliation(s)
- Nafiye Helvaci
- Department of Endocrinology and Metabolism, Hacettepe University School of Medicine, Ankara, Turkey
| | - Serkan Kabacam
- Department of Medical Genetics, Hacettepe University School of Medicine, Ankara, Turkey
| | - Selcuk Dagdelen
- Department of Endocrinology and Metabolism, Hacettepe University School of Medicine, Ankara, Turkey
| | - Incilay Lay
- Department of Medical Biochemistry, Hacettepe University School of Medicine, Ankara, Turkey
| | - Erdem Karabulut
- Department of Biostatistics, Hacettepe University School of Medicine, Ankara, Turkey
| | - Melike Mut
- Department of Neurosurgery, Hacettepe University School of Medicine, Ankara, Turkey
| | - Mehmet Alikasifoglu
- Department of Medical Genetics, Hacettepe University School of Medicine, Ankara, Turkey
| | - Tomris Erbas
- Department of Endocrinology and Metabolism, Hacettepe University School of Medicine, Ankara, Turkey
| |
Collapse
|
158
|
Beckner ME, Conkright WR, Eagle SR, Martin BJ, Sinnott AM, LaGoy AD, Proessl F, Lovalekar M, Jabloner LR, Roma PG, Basner M, Ferrarelli F, Germain A, Flanagan SD, Connaboy C, Nindl BC. Impact of simulated military operational stress on executive function relative to trait resilience, aerobic fitness, and neuroendocrine biomarkers. Physiol Behav 2021; 236:113413. [PMID: 33811909 DOI: 10.1016/j.physbeh.2021.113413] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/11/2021] [Accepted: 03/29/2021] [Indexed: 01/06/2023]
Abstract
PURPOSE To study the impact of 48 h of simulated military operational stress (SMOS) on executive function, in addition to the role of trait resilience (RES) and aerobic fitness (FIT) on executive function performance. Associations between executive function and neuropeptide-Y (NPY), brain-derived neurotropic factor (BDNF), insulin-like growth factor-I (IGF-I), oxytocin, and α-klotho (klotho) were assessed to elucidate potential biomarkers that may contribute to cognitive performance during a multi-factorial stress scenario. METHODS Fifty-four service members (SM) (26.4 ± 5.4 years, 178.0 ± 6.5 cm, 85.2 ± 14.0 kg) completed the 5-day protocol, including daily physical exertion and 48 h of restricted sleep and caloric intake. Each morning subjects completed a fasted blood draw followed by Cognition, a 10-part cognitive test battery assessing executive function. SMs were grouped into tertiles [low (L-), moderate (M-), high (H-)] based on Connor Davidson Resilience Score (RES) and V˙O2peak (FIT). Repeated measures ANOVA were run to analyze the effect of day on cognitive performance and biomarker concentration. Separate two-way mixed ANOVAs were run to determine the interaction of group by day on cognitive function. Friedman test with Bonferroni-corrected pairwise comparisons were used if assumptions for ANOVA were not met. Associations between changes in biomarkers and cognitive performance were analyzed using parametric and non-parametric correlation coefficients. RESULTS SMOS reduced SM vigilance -11.3% (p < 0.001) and working memory -5.6% (p = 0.015), and increased risk propensity +9.5% (p = 0.005). H-RES and H-FIT SMs demonstrated stable vigilance across SMOS (p > 0.05). Vigilance was compromised during SMOS in L- and M-RES (p = 0.007 and p = 0.001, respectively) as well as L- and M-FIT (p = 0.001 and p = 0.031, respectively). SMOS reduced circulating concentrations of α-klotho -7.2% (p = 0.004), NPY -6.4% (p = 0.001), and IGF-I -8.1% (p < 0.001) from baseline through the end of the protocol. BDNF declined -19.2% after the onset of sleep and caloric restriction (p = 0.005) with subsequent recovery within 48 h. Oxytocin remained stable (p > 0.05). Several modest associations between neuroendocrine biomarkers and cognitive performance were identified. CONCLUSION This study demonstrates H-FIT and H-RES may buffer the impact of SMOS on vigilance. SMOS negatively impacted circulating neuroendocrine biomarkers. While BDNF returned to baseline concentrations by the end of the 5 d protocol, NPY, IGF-I, and α-klotho may require a longer recovery period. These data suggest that the military may benefit by training and/or selection processes targeting at augmenting trait resilience and aerobic fitness for increased readiness.
Collapse
Affiliation(s)
- Meaghan E Beckner
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA.
| | - William R Conkright
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shawn R Eagle
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian J Martin
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aaron M Sinnott
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alice D LaGoy
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA; Military Sleep Tactics and Resilience Research Team, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Felix Proessl
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mita Lovalekar
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leslie R Jabloner
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter G Roma
- Behavioral Health & Performance Laboratory, Biomedical Research and Environmental Sciences Division, KBR/NASA Johnson Space Center, Houston, TX, USA
| | - Mathias Basner
- Division of Sleep and Chronobiology, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Fabio Ferrarelli
- Military Sleep Tactics and Resilience Research Team, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anne Germain
- Military Sleep Tactics and Resilience Research Team, Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shawn D Flanagan
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher Connaboy
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bradley C Nindl
- Neuromuscular Research Laboratory/Warrior Human Performance Research Center, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
159
|
Kakar RS, Pastor JV, Moe OW, Ambrosio F, Castaldi D, Sanders LH. Peripheral Klotho and Parkinson's Disease. Mov Disord 2021; 36:1274-1276. [PMID: 33629770 DOI: 10.1002/mds.28530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/25/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Rumit S Kakar
- Old Dominion University Norfolk, Norfolk, Virginia, USA
| | - Johanne V Pastor
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Orson W Moe
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | - Laurie H Sanders
- Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
160
|
Stenhouse C, Halloran KM, Newton MG, Gaddy D, Suva LJ, Bazer FW. Novel mineral regulatory pathways in ovine pregnancy: I. phosphate, klotho signaling, and sodium-dependent phosphate transporters. Biol Reprod 2021; 104:1084-1096. [PMID: 33624764 DOI: 10.1093/biolre/ioab028] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/25/2021] [Accepted: 02/12/2021] [Indexed: 12/30/2022] Open
Abstract
Appropriate mineralization of the fetal skeleton requires an excess of phosphate in the fetus compared to the mother. However, mechanisms for placental phosphate transport are poorly understood. This study aimed to identify phosphate regulatory pathways in ovine endometria and placentae throughout gestation. Suffolk ewes were bred with fertile rams upon visual detection of estrus (Day 0). On Days 9, 12, 17, 30, 70, 90, 110, and 125 of pregnancy (n = 3-14/Day), ewes were euthanized and hysterectomized. Phosphate abundance varied across gestational days in uterine flushings, allantoic fluid, and homogenized endometria and placentae (P < 0.05). The expression of mRNAs for sodium-dependent phosphate transporters (SLC20A1 and SLC20A2) and klotho signaling mediators (FGF7, FGF21, FGF23, FGFR1-4, KL, KLB, ADAM10, and ADAM17) were quantified by qPCR. Day 17 conceptus tissue expressed SLC20A1, SLC20A2, KLB, FGF7, FGF21, FGF23, FGFR1, and FGFR2 mRNAs. Both sodium-dependent phosphate transporters and klotho signaling mediators were expressed in endometria and placentae throughout gestation. Gestational day influenced the expression of SLC20A1, ADAM10, ADAM17, FGF21, FGFR1, and FGFR3 mRNAs in both endometria and placentae (P < 0.05). Gestational day influenced endometrial expression of FGF7 (P < 0.001), and placental expression of FGF23 (P < 0.05). Immunohistochemistry confirmed that both FGF23 and KL proteins were expressed in endometria and placentae throughout gestation. The observed spatiotemporal profile of KL-FGF signaling suggests a potential role in the establishment of pregnancy and regulation of fetal growth. This study provides a platform for further mechanistic investigation into the role for KL-FGF signaling in the regulation of phosphate transport at the ovine maternal-conceptus interface.
Collapse
Affiliation(s)
- Claire Stenhouse
- Departments of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Katherine M Halloran
- Departments of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Makenzie G Newton
- Departments of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Dana Gaddy
- Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Larry J Suva
- Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas, USA
| | - Fuller W Bazer
- Departments of Animal Science, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
161
|
Akasaka-Manya K, Manya H, Nadanaka S, Kitagawa H, Kondo Y, Ishigami A, Endo T. Decreased ADAM17 expression in the lungs of α-Klotho reduced mouse. J Biochem 2021; 167:483-493. [PMID: 31951006 DOI: 10.1093/jb/mvz113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 12/11/2019] [Indexed: 01/26/2023] Open
Abstract
The deficiency of α-Klotho in mice causes phenotypes resembling human age-associated disorders at 3-4 weeks after birth and shows short lifespans of ∼2 months. One of the crucial symptoms is pulmonary emphysema, although α-Klotho is not expressed in the lungs. α-Klotho secreted from the kidneys is probably involved in the pathology of emphysema because kidney-specific knockout mice exhibit emphysematous structural changes. We examined whether any glycan changes in α-Klotho mouse lungs were observed, because α-Klotho is reported to have glycosidase activity. Here, we found the accumulation of heparan sulphate in the microsomal fraction of α-Klotho mouse lungs. Meanwhile, a disintegrin and metalloproteinase 17 (ADAM17) expression was decreased in α-Klotho mice. From these results, it is thought that the increase in heparan sulphate is due to insufficient cleavage of the core protein by ADAM17. Additionally, a reduction in α-Klotho and a decline of ADAM17 were also observed both in normal aged mice and in senescence marker protein-30 (SMP30) knockout mice, a mouse model of premature ageing. Thus, the decrease in ADAM17 is caused by the reduction in α-Klotho. These may be involved in the deterioration of lung function during ageing and may be associated with the pathology of pulmonary emphysema.
Collapse
Affiliation(s)
- Keiko Akasaka-Manya
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Hiroshi Manya
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Satomi Nadanaka
- Laboratory of Biochemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558, Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558, Japan
| | - Yoshitaka Kondo
- Molecular Regulation of Aging, Research Team for Functional Biogerontology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo 173-0015, Japan
| | - Akihito Ishigami
- Molecular Regulation of Aging, Research Team for Functional Biogerontology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo 173-0015, Japan
| | - Tamao Endo
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan
| |
Collapse
|
162
|
Russell DL, Oates JC, Markiewicz M. Association Between the Anti-Aging Gene Klotho and Selected Rheumatologic Autoimmune Diseases. Am J Med Sci 2021; 361:169-175. [PMID: 33349438 PMCID: PMC9741923 DOI: 10.1016/j.amjms.2020.10.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/25/2020] [Accepted: 10/22/2020] [Indexed: 12/14/2022]
Abstract
Klotho long recognized for its role in anti-aging, is potentially implicated in the pathogenesis of rheumatoid arthritis, systemic lupus erythematosus, and systemic sclerosis. Aging of the immune system coincides with the inability of the body to recognize self-antigens, which often leads to autoimmune responses. The role of Klotho in these autoimmune diseases should be of high interest; however, few articles have been published exploring the role of Klotho in the pathogenesis, organ involvement, or clinical manifestation of rheumatoid arthritis, systemic lupus erythematosus, and systemic sclerosis. Herein, we discuss information gathered from peer-reviewed publications to describe the emerging role of Kl in these select rheumatologic autoimmune diseases.
Collapse
Affiliation(s)
| | - Jim C Oates
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina;,Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Margaret Markiewicz
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
163
|
Could α-Klotho Unlock the Key Between Depression and Dementia in the Elderly: from Animal to Human Studies. Mol Neurobiol 2021; 58:2874-2885. [PMID: 33527303 DOI: 10.1007/s12035-021-02313-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 01/25/2021] [Indexed: 10/22/2022]
Abstract
α-Klotho is known for its aging-related functions and is associated with neurodegenerative diseases, accelerated aging, premature morbidity, and mortality. Recent literature suggests that α-Klotho is also involved in the regulation of mental functions, such as cognition and psychosis. While most of studies of α-Klotho are focusing on its anti-aging functions and protective role in dementia, increasing evidence showed many shared symptoms between depression and dementia, while depression has been proposed as the preclinical stage of dementia such as Alzheimer's disease (AD). To see whether and how α-Klotho can be a key biological link between depression and dementia, in this review, we first gathered the evidence on biological distribution and function of α-Klotho in psychiatric functions from animal studies to human clinical investigations with a focus on the regulation of cognition and mood. Then, we discussed and highlighted the potential common underlying mechanisms of α-Klotho between psychiatric diseases and cognitive impairment. Finally, we hypothesized that α-Klotho might serve as a neurobiological link between depression and dementia through the regulation of oxidative stress and inflammation.
Collapse
|
164
|
Xing L, Fang J, Zhu B, Wang L, Chen J, Wang Y, Huang J, Wang H, Yao X. Astragaloside IV protects against podocyte apoptosis by inhibiting oxidative stress via activating PPARγ-Klotho-FoxO1 axis in diabetic nephropathy. Life Sci 2021; 269:119068. [PMID: 33476631 DOI: 10.1016/j.lfs.2021.119068] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/05/2021] [Accepted: 01/13/2021] [Indexed: 01/07/2023]
Abstract
AIMS Podocyte apoptosis plays an important role in the pathogenesis of diabetic nephropathy (DN). Astragaloside IV (AS-IV) has been shown to protect against podocyte apoptosis. Here we aim to investigate the mechanism responsible for the protective effects of AS-IV. MAIN METHODS Diabetic db/db mice and high glucose (HG)-cultured podocytes were treated with AS-IV. Renal function and histopathological changes were measured to evaluate the therapeutic effects of AS-IV against DN. Adenovirus-mediated Klotho overexpression, Klotho siRNA, and PPARγ inhibitor were applied in vitro to investigate the potential mechanism. The expression levels of mRNA and proteins were analyzed by qRT-PCR, western blot or immunofluorescence. Intracellular ROS and mitochondrial superoxide were detected by DHE and MitoSOx Red, respectively. Cell apoptosis was evaluated by TUNEL staining and flow cytometry. KEY FINDINGS AS-IV improved renal function and ameliorated podocyte injury in db/db mice accompanied with enhanced Klotho expression in glomerular podocytes. In vitro, AS-IV inhibited HG-induced podocyte apoptosis and restored HG-inhibited Klotho expression, whereas Klotho knockdown abrogated the anti-apoptosis action of AS-IV. Further study showed that adenovirus-mediated Klotho overexpression enhanced Forkhead transcription factor O1 (FoxO1)-dependent antioxidant activity and attenuated HG-evoked oxidative stress and apoptosis. AS-IV prevented HG-induced FoxO1 inhibition and oxidative stress, whereas Klotho knockdown reversed these effects. Cotreatment with PPARγ inhibitor T0070907 abolished AS-IV-induced Klotho expression and anti-apoptosis action. SIGNIFICANCE These data suggested that AS-IV attenuated podocyte apoptosis presumably by inhibiting oxidative stress via activating PPARγ-Klotho-FoxO1 signaling pathway, thereby ameliorating DN. This study provided new insights into the molecular mechanisms of AS-IV against DN.
Collapse
Affiliation(s)
- Lina Xing
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Ji Fang
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Bingbing Zhu
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Li Wang
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Junliang Chen
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Yunman Wang
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Jiebo Huang
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Hao Wang
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| | - Xingmei Yao
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| |
Collapse
|
165
|
Tempaku PF, D'Almeida V, da Silva SMA, Andersen ML, Belangero SI, Tufik S. Klotho genetic variants mediate the association between obstructive sleep apnea and short telomere length. Sleep Med 2021; 83:210-213. [PMID: 34049038 DOI: 10.1016/j.sleep.2021.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/30/2020] [Accepted: 01/12/2021] [Indexed: 11/30/2022]
Abstract
The core features of obstructive sleep apnea (OSA) can potentially contribute to the acceleration of telomere shortening mechanisms. Other factor associated with telomeres is Klotho gene as it can negatively regulates telomerase activity. Noteworthy, KLOTHO protein level has recently been associated with OSA. In this sense, it was plausible to hypothesize that OSA would be associated with short telomere length and those with OSA plus risk single nucleotide polymorphisms (SNPs) in Klotho gene would present even shorter telomere length. As part of the EPISONO cohort, 1042 individuals answered questionnaires, underwent polysomnography and had blood collected for DNA extraction. OSA was defined according to AHI≥ 15 events/hour. Leukocyte telomere length (LTL) was measured through real-time polymerase chain reaction (qPCR) and Klotho SNPs were genotyped by array. Mediation analyses considered the presence of SNPs in Klotho gene and how this interaction can affect OSA and its consequence in telomere length. All the analyses were corrected for multiple comparisons. LTL was significantly shorter in OSA compared to controls in a severity-dependent manner (B = 0.055; CI = 0.007-0.102; p = 0.02). Among the 43 Klotho SNPs analyzed, we observed that 4 SNPs (rs525014, rs7982726, rs685417 and rs9563124) significantly mediated the association between OSA and short LTL. Klotho gene opens a new venue in OSA research since it can contribute in the increase of knowledge of the mechanisms involved in the consequences of short telomeres in individuals with OSA.
Collapse
Affiliation(s)
- Priscila Farias Tempaku
- Departamento de Psicobiologia and Morfologia e Genética, Universidade Federal de São Paulo, Brazil
| | - Vânia D'Almeida
- Departamento de Psicobiologia and Morfologia e Genética, Universidade Federal de São Paulo, Brazil
| | | | - Monica Levy Andersen
- Departamento de Psicobiologia and Morfologia e Genética, Universidade Federal de São Paulo, Brazil
| | | | - Sergio Tufik
- Departamento de Psicobiologia and Morfologia e Genética, Universidade Federal de São Paulo, Brazil.
| |
Collapse
|
166
|
Ewendt F, Feger M, Föller M. Role of Fibroblast Growth Factor 23 (FGF23) and αKlotho in Cancer. Front Cell Dev Biol 2021; 8:601006. [PMID: 33520985 PMCID: PMC7841205 DOI: 10.3389/fcell.2020.601006] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/15/2020] [Indexed: 12/16/2022] Open
Abstract
Together with fibroblast growth factors (FGFs) 19 and 21, FGF23 is an endocrine member of the family of FGFs. Mainly secreted by bone cells, FGF23 acts as a hormone on the kidney, stimulating phosphate excretion and suppressing formation of 1,25(OH)2D3, active vitamin D. These effects are dependent on transmembrane protein αKlotho, which enhances the binding affinity of FGF23 for FGF receptors (FGFR). Locally produced FGF23 in other tissues including liver or heart exerts further paracrine effects without involvement of αKlotho. Soluble Klotho (sKL) is an endocrine factor that is cleaved off of transmembrane Klotho or generated by alternative splicing and regulates membrane channels, transporters, and intracellular signaling including insulin growth factor 1 (IGF-1) and Wnt pathways, signaling cascades highly relevant for tumor progression. In mice, lack of FGF23 or αKlotho results in derangement of phosphate metabolism and a syndrome of rapid aging with abnormalities affecting most organs and a very short life span. Conversely, overexpression of anti-aging factor αKlotho results in a profound elongation of life span. Accumulating evidence suggests a major role of αKlotho as a tumor suppressor, at least in part by inhibiting IGF-1 and Wnt/β-catenin signaling. Hence, in many malignancies, higher αKlotho expression or activity is associated with a more favorable outcome. Moreover, also FGF23 and phosphate have been revealed to be factors relevant in cancer. FGF23 is particularly significant for those forms of cancer primarily affecting bone (e.g., multiple myeloma) or characterized by bone metastasis. This review summarizes the current knowledge of the significance of FGF23 and αKlotho for tumor cell signaling, biology, and clinically relevant parameters in different forms of cancer.
Collapse
Affiliation(s)
- Franz Ewendt
- Department of Nutritional Physiology, Institute of Agricultural and Nutritional Sciences, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - Martina Feger
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Michael Föller
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
167
|
Lee S, Jung MH, Song K, Jin JX, Taweechaipaisankul A, Kim GA, Oh HJ, Koo OJ, Park SC, Lee BC. Failure to maintain full-term pregnancies in pig carrying klotho monoallelic knockout fetuses. BMC Biotechnol 2021; 21:1. [PMID: 33413301 PMCID: PMC7791653 DOI: 10.1186/s12896-020-00660-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 12/04/2020] [Indexed: 12/30/2022] Open
Abstract
Background Small animals that show a deficiency in klotho exhibit extremely shortened life span with multiple aging-like phenotypes. However, limited information is available on the function of klotho in large animals such as pigs. Results In an attempt to produce klotho knockout pigs, an sgRNA specific for klotho (targeting exon 3) was designed and Cas9-sgRNA ribonucleoproteins were transfected into porcine fibroblasts. Transfected fibroblasts were cultured for one to 2 days and then directly used for nuclear transfer without selection. The cloned embryos were cultured in vitro for 7 days and analyzed to detect modifications of the klotho gene by both T7E1 and deep sequencing analysis. Modification succeeded in 13 of 20 blastocysts (65%), 8 of which (40.0%) were monoallelic modifications and 5 (25.0%) were biallelic modifications. Based on high mutation rates in blastocysts, we transferred the cloned embryos to 5 recipient pigs; 1 recipient was pregnant and 16 fetuses were recovered at Day 28 post transfer. Of the 16 fetuses, 9 were resorbing and 7 were viable. Four of 9 (44.4%) resorbing fetuses and 3 of the 7 (42.9%) viable fetuses had monoallelic modifications. Thus, 3 klotho monoallelic knockout cell lines were established by primary culture. A total of 2088 cloned embryos reconstructed with 2 frame-shifted cell lines were transferred to 11 synchronized recipients. Of the recipients, 7 of 11 eleven (63.6%) became pregnant. However, none of the pregnancies was maintained to term. To discover why klotho monoallelic knockout fetuses were aborted, expression of aging- and apoptosis-related genes and klotho protein in placentas from klotho monoallelic knockout and wild-type fetuses was investigated. Placentas from klotho monoallelic knockout fetuses showed negatively changed expression of aging- and apoptosis-related genes with lower relative expression of klotho protein. These results indicated that the reason why klotho monoallelic knockout fetuses were not maintained to term was possibly due to decreased klotho expression in placentas, negatively affecting aging- and apoptosis-related genes. Conclusions Klotho monoallelic knockout porcine fetal fibroblasts were successfully established. However, pigs carrying klotho monoallelic knockout fetuses failed to maintain full-term pregnancy and a decrease in klotho expression in placenta likely leads to pregnancy loss. Supplementary Information The online version contains supplementary material available at 10.1186/s12896-020-00660-9.
Collapse
Affiliation(s)
- Sanghoon Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | | | - Kilyoung Song
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jun-Xue Jin
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Anukul Taweechaipaisankul
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Geon A Kim
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon, Republic of Korea
| | - Hyun Ju Oh
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | | | - Se Chang Park
- Laboratory of Aquatic Biomedicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Byeong Chun Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
168
|
Câmara AB, Brandão IA. The relationship between vitamin D deficiency and oxidative stress can be independent of age and gender. INT J VITAM NUTR RES 2021; 91:108-123. [DOI: 10.1024/0300-9831/a000614] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Abstract. The active vitamin D (1,25(OH)2D) acts through a nuclear receptor to perform several functions in cellular metabolism. 1,25(OH)2D participates directly in calcium homeostasis, regulates the immune system, nervous system, blood pressure, insulin secretion, among others. Vitamin D deficiency could also be associated with several diseases and increased cellular oxidative damage. The present study aimed to investigate whether lipid peroxidation and/or protein oxidation are affected by vitamin D deficiency and whether sunlight exposure/diet, gender, and age might influence this relationship. Vitamin D concentrations were obtained from the Heart Hospital database and a questionnaire was applied among the 212 participants. We used the inactive vitamin D (25(OH)2) in the analyses since 1,25(OH)2D has a short half-life and a low blood concentration. Lipid peroxidation and protein oxidation analyses were performed using spectrophotometry. Multivariate analyses suggested the participation of vitamin D deficiency (<30 ng/mL) and sunlight/diet in oxidative stress (p <0.05; R2 MDA: 0.562; R2 CG: 0.429). Multiple linear regression test show that the age and gender of patients are not interfering in the analyses (p>0.05). Therefore, we suggest that the relationship between vitamin D deficiency and oxidative stress can be independent of age and gender.
Collapse
Affiliation(s)
- Alice Barros Câmara
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal/RN, Brazil
| | - Igor Augusto Brandão
- Metrópole Digital Institute, Federal University of Rio Grande do Norte, Natal/RN, Brazil
| |
Collapse
|
169
|
Dias GP, Murphy T, Stangl D, Ahmet S, Morisse B, Nix A, Aimone LJ, Aimone JB, Kuro-O M, Gage FH, Thuret S. Intermittent fasting enhances long-term memory consolidation, adult hippocampal neurogenesis, and expression of longevity gene Klotho. Mol Psychiatry 2021; 26:6365-6379. [PMID: 34031536 PMCID: PMC8760057 DOI: 10.1038/s41380-021-01102-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 03/19/2021] [Accepted: 04/06/2021] [Indexed: 02/03/2023]
Abstract
Daily calorie restriction (CR) and intermittent fasting (IF) enhance longevity and cognition but the effects and mechanisms that differentiate these two paradigms are unknown. We examined whether IF in the form of every-other-day feeding enhances cognition and adult hippocampal neurogenesis (AHN) when compared to a matched 10% daily CR intake and ad libitum conditions. After 3 months under IF, female C57BL6 mice exhibited improved long-term memory retention. IF increased the number of BrdU-labeled cells and neuroblasts in the hippocampus, and microarray analysis revealed that the longevity gene Klotho (Kl) was upregulated in the hippocampus by IF only. Furthermore, we found that downregulating Kl in human hippocampal progenitor cells led to decreased neurogenesis, whereas Kl overexpression increased neurogenesis. Finally, histological analysis of Kl knockout mice brains revealed that Kl is required for AHN, particularly in the dorsal hippocampus. These data suggest that IF is superior to 10% CR in enhancing memory and identifies Kl as a novel candidate molecule that regulates the effects of IF on cognition likely via AHN enhancement.
Collapse
Affiliation(s)
- Gisele Pereira Dias
- grid.13097.3c0000 0001 2322 6764Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Tytus Murphy
- grid.13097.3c0000 0001 2322 6764Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Doris Stangl
- grid.13097.3c0000 0001 2322 6764Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Selda Ahmet
- grid.13097.3c0000 0001 2322 6764Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Benjamin Morisse
- grid.13097.3c0000 0001 2322 6764Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Alina Nix
- grid.13097.3c0000 0001 2322 6764Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Lindsey J. Aimone
- grid.250671.70000 0001 0662 7144Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA USA
| | - James B. Aimone
- grid.474520.00000000121519272Center for Computing Research, Sandia National Laboratories, Albuquerque, NM USA
| | - Makoto Kuro-O
- grid.410804.90000000123090000Division of Anti-Ageing Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Fred H. Gage
- grid.250671.70000 0001 0662 7144Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA USA
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK. .,Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
170
|
Gao X, Sun Z, Ma G, Li Y, Liu M, Zhang G, Xu H, Gao Y, Zhou J, Deng Q, Li R. Reduced Plasma Levels of α-Klotho and Their Correlation With Klotho Polymorphisms in Elderly Patients With Major Depressive Disorders. Front Psychiatry 2021; 12:682691. [PMID: 34721095 PMCID: PMC8548667 DOI: 10.3389/fpsyt.2021.682691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Recent literature suggests that α-Klotho, a widely recognized anti-aging protein, is involved in longevity as well as in many diseases, including Alzheimer's disease, and depression. Although the Klotho gene encodes α-Klotho, a single transmembrane protein with intracellular and extracellular domains, the relationship between Klotho gene polymorphism and circulating α-Klotho levels in patients with major depressive disorder (MDD) is not clear. Methods: A total of 144 MDD patients and 112 age-matched healthy controls were included in this study. The Klotho genetic polymorphisms (rs9536314, rs9527025, and rs9315202) and plasma α-Klotho levels were measured by PCR and ELISA, respectively. The severity of depressive symptoms was estimated using the Hamilton Depression Scale (HAMD). Results: We found a significantly lower level of plasma α-Klotho in the MDD patients than in controls. Among them, only elderly MDD patients (first episode) showed significantly lower α-Klotho levels than the age-matched controls, while elderly recurrent and young MDD patients showed no difference in plasma α-Klotho levels from age-matched controls. The young MDD group showed a significantly earlier onset age, higher plasma α-Klotho levels, and lower HAMD scores than those in the elderly MDD group. While the plasma α-Klotho levels were higher in rs9315202 T alleles carrier regardless age or sex, the rs9315202 T allele was negatively correlated with disease severity only in the elderly MDD patients. Conclusion: The results of our study showed that only elderly MDD patients showed a decrease in plasma α-Klotho levels along with an increase in disease severity as well as an association with the number of rs9315202 T alleles, and not young MDD patients compared to age-matched controls. Our data suggest that circulating α-Klotho levels combined with Klotho genetic polymorphisms are important in elderly MDD patients, particularly carriers of the Klotho gene rs9315202 T allele.
Collapse
Affiliation(s)
- Xiang Gao
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Zuoli Sun
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Guangwei Ma
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Yuhong Li
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Min Liu
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Guofu Zhang
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Hong Xu
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Yane Gao
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Jixuan Zhou
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Qi Deng
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Rena Li
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
171
|
Neyra JA, Hu MC, Moe OW. Klotho in Clinical Nephrology: Diagnostic and Therapeutic Implications. Clin J Am Soc Nephrol 2020; 16:162-176. [PMID: 32699047 PMCID: PMC7792642 DOI: 10.2215/cjn.02840320] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
αKlotho (called Klotho here) is a membrane protein that serves as the coreceptor for the circulating hormone fibroblast growth factor 23 (FGF23). Klotho is also cleaved and released as a circulating substance originating primarily from the kidney and exerts a myriad of housekeeping functions in just about every organ. The vital role of Klotho is shown by the multiorgan failure with genetic deletion in rodents, with certain features reminiscent of human disease. The most common causes of systemic Klotho deficiency are AKI and CKD. Preclinical data on Klotho biology have advanced considerably and demonstrated its potential diagnostic and therapeutic value; however, multiple knowledge gaps exist in the regulation of Klotho expression, release, and metabolism; its target organs; and mechanisms of action. In the translational and clinical fronts, progress has been more modest. Nonetheless, Klotho has potential clinical applications in the diagnosis of AKI and CKD, in prognosis of progression and extrarenal complications, and finally, as replacement therapy for systemic Klotho deficiency. The overall effect of Klotho in clinical nephrology requires further technical advances and additional large prospective human studies.
Collapse
Affiliation(s)
- Javier A. Neyra
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Nephrology, Bone and Mineral Metabolism, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Orson W. Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
172
|
Cioffi F, Adam RHI, Broersen K. Molecular Mechanisms and Genetics of Oxidative Stress in Alzheimer's Disease. J Alzheimers Dis 2020; 72:981-1017. [PMID: 31744008 PMCID: PMC6971833 DOI: 10.3233/jad-190863] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alzheimer’s disease is the most common neurodegenerative disorder that can cause dementia in elderly over 60 years of age. One of the disease hallmarks is oxidative stress which interconnects with other processes such as amyloid-β deposition, tau hyperphosphorylation, and tangle formation. This review discusses current thoughts on molecular mechanisms that may relate oxidative stress to Alzheimer’s disease and identifies genetic factors observed from in vitro, in vivo, and clinical studies that may be associated with Alzheimer’s disease-related oxidative stress.
Collapse
Affiliation(s)
- Federica Cioffi
- Nanobiophysics Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Rayan Hassan Ibrahim Adam
- Nanobiophysics Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Kerensa Broersen
- Applied Stem Cell Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| |
Collapse
|
173
|
Lanzani C, Citterio L, Vezzoli G. Klotho: a link between cardiovascular and non-cardiovascular mortality. Clin Kidney J 2020; 13:926-932. [PMID: 33391735 PMCID: PMC7769552 DOI: 10.1093/ckj/sfaa100] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/08/2020] [Indexed: 12/22/2022] Open
Abstract
Klotho is a membrane-bound protein acting as an obligatory coreceptor for fibroblast growth factor 23 (FGF23) in the kidney and parathyroid glands. The extracellular portion of its molecule may be cleaved and released into the blood and produces multiple endocrine effects. Klotho exerts anti-inflammatory and antioxidative activities that may explain its ageing suppression effects evidenced in mice; it also modulates mineral metabolism and FGF23 activities and limits their negative impact on cardiovascular system. Clinical studies have found that circulating Klotho is associated with myocardial hypertrophy, coronary artery disease and stroke and may also be involved in the pathogenesis of salt-sensitive hypertension with a mechanism sustained by inflammatory cytokines. As a consequence, patients maintaining high serum levels of Klotho not only show decreased cardiovascular mortality but also non-cardiovascular mortality. Klotho genetic polymorphisms may influence these clinical relationships and predict cardiovascular risk; rs9536314 was the polymorphism most frequently involved in these associations. These findings suggest that Klotho and its genetic polymorphisms may represent a bridge between inflammation, salt sensitivity, hypertension and mortality. This may be particularly relevant in patients with chronic kidney disease who have decreased Klotho levels in tissues and blood.
Collapse
Affiliation(s)
- Chiara Lanzani
- Nephrology and Dialysis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Genomics of Renal Diseases and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorena Citterio
- Genomics of Renal Diseases and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe Vezzoli
- Nephrology and Dialysis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Genomics of Renal Diseases and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
174
|
Lodjak J, Verhulst S. Insulin-like growth factor 1 of wild vertebrates in a life-history context. Mol Cell Endocrinol 2020; 518:110978. [PMID: 32798584 DOI: 10.1016/j.mce.2020.110978] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022]
Abstract
Broad variation in intra- and interspecific life-history traits is largely shaped by resource limitation and the ensuing allocation trade-offs that animals are forced to make. Insulin-like growth factor 1 (IGF-1), a growth-hormone-dependent peptide, may be a key player in the regulation of allocation processes. In laboratory animals, the effects of IGF-1 on growth- and development (positive), reproduction (positive), and longevity (negative) are well established. We here review the evidence on these effects in wild vertebrates, where animals are more likely to face resource limitation and other challenges. We point out the similarities and dissimilarities in patterns of IGF-1 functions obtained in these two different study settings and discuss the knowledge we need to develop a comprehensive picture of the role of IGF-1 in mediating life-history variation of wild vertebrates.
Collapse
Affiliation(s)
- Jaanis Lodjak
- Department of Zoology, Institute of Ecology and Earth Sciences, University of Tartu, 46 Vanemuise Street, Tartu, 51014, Estonia; Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, Netherlands.
| | - Simon Verhulst
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, Netherlands
| |
Collapse
|
175
|
Xu Z, Zheng S, Feng X, Cai C, Ye X, Liu P. Klotho gene improves oxidative stress injury after myocardial infarction. Exp Ther Med 2020; 21:52. [PMID: 33273980 PMCID: PMC7706392 DOI: 10.3892/etm.2020.9484] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 04/07/2020] [Indexed: 11/17/2022] Open
Abstract
The aim of the present study was to investigate the effects and mechanisms of the Klotho gene in oxidative stress injury after myocardial infarction. Sprague-Dawley rats were divided into five groups (sham, model, pDC316, LY294002, and pDC316-Klotho). Subsequently, the superoxide dismutase (SOD), glutathione (GSH), and malondialdehyde (MDA) concentrations were measured in myocardial tissues. Additionally, pathological differences among the groups were evaluated using hematoxylin and eosin and Masson's trichrome staining. Apoptosis was assayed by terminal deoxynucleotidyl transferase 2'-deoxyuridine-5'-triphosphate nick end-labeling assay, evaluated Klotho protein expression by immunohistochemical assay, and assessed Nrf 2 and ARE protein expressions using western blotting assay. As compared with in the sham group, the SOD, MDA, and GSH concentrations were significantly deteriorated (P<0.001, respectively); cardiomyocyte apoptosis index values were significantly increased (P<0.001); Klotho protein expression was significantly depressed; and Nrf-2 and ARE protein expressions were significantly (P<0.001, respectively) in the model and pDC316 groups. However, with Klotho supplementation by pDC316 transfection, as compared with in the model group, the SOD, MDA, and GSH concentrations were significantly improved (P<0.001, respectively); the cardiomyocyte apoptosis index values were significantly suppressed (P<0.001); and the pathology was improved. Further, the Klotho protein expression of the pDC316-Klotho group was significantly upregulated and the Nrf-2 and ARE proteins expressions of the LY294002 and pDC316-Klotho groups were significantly suppressed. Klotho overexpression improved findings of oxidative stress injury after myocardial infarction.
Collapse
Affiliation(s)
- Zhuofan Xu
- Department of Internal Medicine-Cardiovascular, Guangzhou 12th People's Hospital, Guangzhou, Guangdong 510620, P.R. China
| | - Shaoxin Zheng
- Department of Internal Medicine-Cardiovascular, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510200, P.R. China
| | - Xiaoqian Feng
- Department of Internal Medicine-Cardiovascular, Guangzhou 12th People's Hospital, Guangzhou, Guangdong 510620, P.R. China
| | - Chengzhe Cai
- Department of Internal Medicine-Cardiovascular, Guangzhou 12th People's Hospital, Guangzhou, Guangdong 510620, P.R. China
| | - Xianqu Ye
- Department of Internal Medicine-Cardiovascular, Guangzhou 12th People's Hospital, Guangzhou, Guangdong 510620, P.R. China
| | - Pingfang Liu
- Department of Internal Medicine-Cardiovascular, Guangzhou 12th People's Hospital, Guangzhou, Guangdong 510620, P.R. China
| |
Collapse
|
176
|
Xu X, Hao Y, Zhong Q, Hang J, Zhao Y, Qiao J. Low KLOTHO level related to aging is associated with diminished ovarian reserve. Fertil Steril 2020; 114:1250-1255. [PMID: 33153705 DOI: 10.1016/j.fertnstert.2020.06.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To explore the relationship between KLOTHO expression and diminished ovarian reserve (DOR). DESIGN A case-control study. SETTING Reproductive medicine center. PATIENT(S) A total of 157 patients with DOR and 159 control women were recruited from the Centre of Reproductive Medicine, Peking University Third Hospital. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) The granulosa cells were isolated from follicular fluid after oocyte retrieval, and the KLOTHO level of granulosa cell was measured using a modified quantitative polymerase chain reaction technique. The serum KLOTHO level was measured by solid-phase sandwich enzyme-linked immunosorbent assay. RESULT(S) In both granulosa cells and serum derived from women with DOR, KLOTHO expressions were significantly lower compared with normal ovarian reserve controls. Moreover, KLOTHO expression diminished with advancing age. CONCLUSION(S) Diminished KLOTHO expression was associated with DOR. Further longitudinal studies in a similar population accompanying disease progression and mechanism exploration are needed to substantiate the rules of KLOTHO in reproductive aging.
Collapse
Affiliation(s)
- Xiaofei Xu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, People's Republic of China; National Clinical Research Center for Obstetrics and Gynecology, Beijing, People's Republic of China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, People's Republic of China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, People's Republic of China
| | - Yongxiu Hao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, People's Republic of China; National Clinical Research Center for Obstetrics and Gynecology, Beijing, People's Republic of China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, People's Republic of China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, People's Republic of China
| | - Qihang Zhong
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People's Republic of China
| | - Jing Hang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, People's Republic of China; National Clinical Research Center for Obstetrics and Gynecology, Beijing, People's Republic of China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, People's Republic of China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, People's Republic of China
| | - Yue Zhao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, People's Republic of China; National Clinical Research Center for Obstetrics and Gynecology, Beijing, People's Republic of China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, People's Republic of China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, People's Republic of China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, People's Republic of China; National Clinical Research Center for Obstetrics and Gynecology, Beijing, People's Republic of China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, People's Republic of China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, People's Republic of China; Beijing Advanced Innovation Center for Genomics, Beijing, People's Republic of China; Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Beijing, People's Republic of China.
| |
Collapse
|
177
|
Kale A, Sankrityayan H, Anders HJ, Gaikwad AB. Epigenetic and non-epigenetic regulation of Klotho in kidney disease. Life Sci 2020; 264:118644. [PMID: 33141039 DOI: 10.1016/j.lfs.2020.118644] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022]
Abstract
Klotho is a novel renoprotective anti-aging protein available in membrane-bound or soluble form. Klotho is expressed in brain, pancreas, and other solid organs but shows highest expression levels in the kidney. Klotho sustains normal kidney physiology but Klotho regulation also contributes to the progression of kidney disease. Systemic and intrarenal levels of Klotho fall drastically during acute kidney injury, kidney fibrosis, diabetic nephropathy, and other forms of chronic kidney disease, etc. Moreover, exogenous supplementation or overexpression of endogenous Klotho attenuates kidney disease. The regulation of endogenous Klotho expression involves epigenetic as well as non-epigenetic mechanisms. The epigenetic modifications such as DNA methylation, post-translational histone modifications, miRNAs regulate the change in Klotho expression in kidney disease. Non-epigenetic mechanisms such as ER stress, Wnt signaling, activation of the renin angiotensin system (RAS), excessive reactive oxygen species and cytokine generation, albumin overload, and PPAR-γ signaling also contribute to Klotho regulation. Evolving evidence highlight the capacity of natural products to regulate Klotho expression in kidney disease. All these preclinical data suggest that Klotho could be a novel biomarker as well as therapeutic target. Here we review the different mechanisms of Klotho regulation in the context of Klotho as a biomarker and potential therapeutic agent.
Collapse
Affiliation(s)
- Ajinath Kale
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Himanshu Sankrityayan
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Internal Medicine IV, University Hospital of the Ludwig Maximilians University Munich, 80336 Munich, Germany
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|
178
|
Olejnik A, Krzywonos-Zawadzka A, Banaszkiewicz M, Bil-Lula I. Ameliorating Effect of Klotho Protein on Rat Heart during I/R Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6427284. [PMID: 33123314 PMCID: PMC7586150 DOI: 10.1155/2020/6427284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/22/2020] [Accepted: 09/29/2020] [Indexed: 01/08/2023]
Abstract
An essential procedure for the treatment of myocardial infarction is restoration of blood flow in the obstructed infarct artery, which may cause ischaemia/reperfusion (I/R) injury. Heart I/R injury manifests in oxidative stress, metabolic and morphological disorders, or cardiac contractile dysfunction. Klotho protein was found to be produced in the heart tissue and participate in antioxidation or ion homeostasis. The aim of this study was to examine an influence of Klotho protein on the heart subjected to I/R injury. Wistar rats served as a surrogate heart model ex vivo. Rat hearts perfused using the Langendorff method were subjected to global no-flow ischaemia, and isolated rat cardiomyocytes underwent chemical I/R in vitro, with or without recombinant Klotho protein administration. Haemodynamic parameters of heart function, cell contractility, markers of I/R injury and oxidative stress, and the level of contractile proteins such as myosin light chain 1 (MLC1) and troponin I (TnI) were measured. The treatment of hearts subjected to I/R injury with Klotho protein resulted in a recovery of heart mechanical function and ameliorated myocyte contractility. This improvement was associated with decreased tissue injury, enhanced antioxidant capacity, and reduced release of MLC1 and TnI. The present research showed the contribution of Klotho to cardioprevention during I/R. Thus, Klotho protein may support the protection from I/R injury and prevention of contractile dysfunction in the rat heart.
Collapse
Affiliation(s)
- Agnieszka Olejnik
- Division of Clinical Chemistry and Laboratory Hematology, Department of Medical Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A St., 50-556 Wroclaw, Poland
| | - Anna Krzywonos-Zawadzka
- Division of Clinical Chemistry and Laboratory Hematology, Department of Medical Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A St., 50-556 Wroclaw, Poland
| | - Marta Banaszkiewicz
- Division of Clinical Chemistry and Laboratory Hematology, Department of Medical Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A St., 50-556 Wroclaw, Poland
| | - Iwona Bil-Lula
- Division of Clinical Chemistry and Laboratory Hematology, Department of Medical Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A St., 50-556 Wroclaw, Poland
| |
Collapse
|
179
|
Ma Z, Liu J, Li J, Jiang H, Kong J. Klotho Levels are Decreased and Associated with Enhanced Oxidative Stress and Inflammation in the Aqueous Humor in Patients with Exudative Age-related Macular Degeneration. Ocul Immunol Inflamm 2020; 30:630-637. [PMID: 33048602 DOI: 10.1080/09273948.2020.1828488] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE To evaluate anti-aging protein klotho levels in the aqueous humor and its association with oxidative stress and inflammation in patients with age-related macular degeneration (AMD). METHODS Levels of klotho, oxidative, and antioxidative stress markers, and proinflammatory and anti-inflammatory markers in the aqueous humor from 28 patients with exudative AMD and 35 age-matched controls were measured. RESULTS Patients with AMD had lower levels of klotho, which were negatively correlated with macular lesion size. Patients with AMD also exhibited increased levels of 8-hydroxy-2'-deoxyguanosine (8-OHdG) and interleukin (IL)-6 but not tumor necrosis factor‑α, and decreased levels of total antioxidant status (TAS) and IL-10. Moreover, levels of klotho were negatively correlated with levels of 8-OHdG and IL-6, but positively correlated with levels of TSA and IL-10. CONCLUSION Klotho levels in the aqueous humor are decreased and associated with oxidative stress and inflammation in patients with exudative AMD.
Collapse
Affiliation(s)
- Zhongxu Ma
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Vision Science, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Jingjing Liu
- Provincial Key Laboratory of Lens Research, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jing Li
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Vision Science, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Hao Jiang
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Vision Science, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Jun Kong
- Provincial Key Laboratory of Lens Research, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
180
|
Effects of Klotho supplementation on hyperoxia-induced renal injury in a rodent model of postnatal nephrogenesis. Pediatr Res 2020; 88:565-570. [PMID: 32059229 PMCID: PMC8226112 DOI: 10.1038/s41390-020-0803-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/18/2019] [Accepted: 02/01/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND Hyperoxia (HO) causes kidney injury in preterm infants; however, whether these effects are modifiable is unknown. We hypothesized that administration of exogenous soluble Klotho, a kidney-derived antioxidant, would attenuate HO-induced kidney injury during postnatal nephrogenesis in rats. METHODS Sprague Dawley neonatal rats assigned to normoxia (21% O2) or HO (85% O2) groups from postnatal day (P) 1 to 21 were randomly assigned to receive alternate day intraperitoneal injections of recombinant Klotho or placebo for 3 weeks. They were recovered in normoxia for an additional 3 weeks and sacrificed at 6 weeks. Renal artery resistance and pulsatility indices, tubular injury scores, glomerular area, and renal antioxidant capacity were assessed. RESULTS Rodents exposed to HO during postnatal nephrogenesis had reduced kidney Klotho expression, glomerulomegaly, and higher tubular injury scores. Exogenous Klotho administration improved renal perfusion as indicated by decreases in both resistance and pulsatility indices and increased antioxidant enzyme expression. CONCLUSIONS HO exposure during postnatal nephrogenesis in rodents results in a decline in kidney Klotho expression, decreased renal perfusion, enlarged glomerular size, and tubular injury. The exogenous administration of Klotho attenuated HO-induced kidney injury and augmented antioxidant capacity.
Collapse
|
181
|
Chen CC, Chang ZY, Tsai FJ, Chen SY. Resveratrol Pretreatment Ameliorates Concanavalin A-Induced Advanced Renal Glomerulosclerosis in Aged Mice through Upregulation of Sirtuin 1-Mediated Klotho Expression. Int J Mol Sci 2020; 21:ijms21186766. [PMID: 32942691 PMCID: PMC7554923 DOI: 10.3390/ijms21186766] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Abstract
Aging kidneys are characterized by an increased vulnerability to glomerulosclerosis and a measurable decline in renal function. Evidence suggests that renal and systemic klotho and sirtuin 1 (SIRT1) deficiencies worsen kidney damage induced by exogenous stresses. The aim of this study was to explore whether resveratrol would attenuate concanavalin A (Con A)-induced renal oxidative stress and advanced glomerulosclerosis in aged mice. Aged male C57BL/6 mice were treated orally with resveratrol (30 mg/kg) seven times (12 h intervals) prior to the administration of a single tail-vein injection of Con A (20 mg/kg). The plasma and urinary levels of kidney damage markers were evaluated. The kidney histopathology, renal parameters, and oxidative stress levels were measured. Furthermore, klotho was downregulated in mouse kidney mesangial cells that were pretreated with 25 µM resveratrol followed by 20 µg/mL Con A. The urinary albumin/creatinine ratio, blood urea nitrogen, kidney mesangial matrix expansion, tubulointerstitial fibrosis, and renal levels of α-smooth muscle actin, transforming growth factor beta, fibronectin, procollagen III propeptide, and collagen type I significantly increased in Con A-treated aged mice. Aged mice kidneys also showed markedly increased levels of 8-hydroxydeoxyguanosine (8-OH-dG) and reactive oxygen species (ROS), with reduced superoxide dismutase activity and levels of glutathione, klotho, and SIRT1 after Con A challenge. Furthermore, in kidney mesangial cells, klotho silencing abolished the effects of resveratrol on the Con A-mediated elevation of the indices of oxidative stress and the expression of glomerulosclerosis-related factors. These findings suggest that resveratrol protects against Con A-induced advanced glomerulosclerosis in aged mice, ameliorating renal oxidative stress via the SIRT1-mediated klotho expression.
Collapse
Affiliation(s)
- Chin-Chang Chen
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung 204, Taiwan; (C.-C.C.); (Z.-Y.C.)
- Department of Anatomy, School of Medicine, China Medical University, Taichung 404, Taiwan
| | - Zi-Yu Chang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung 204, Taiwan; (C.-C.C.); (Z.-Y.C.)
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan;
- Genetics Center, Medical Research, China Medical University Hospital, Taichung 404, Taiwan
- Department of Medical Genetics, China Medical University Hospital, Taichung 404, Taiwan
| | - Shih-Yin Chen
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan;
- Genetics Center, Medical Research, China Medical University Hospital, Taichung 404, Taiwan
- Correspondence:
| |
Collapse
|
182
|
Kamal A, Salama M, Kamal A, Mohsen A, Siam I. Klotho (rs1207568 and rs564481) gene variants and colorectal cancer risk. TURKISH JOURNAL OF GASTROENTEROLOGY 2020; 31:497-502. [PMID: 32897222 DOI: 10.5152/tjg.2020.19235] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND/AIMS Whereas colorectal cancer (CRC) is the third most common cancer worldwide, klotho gene has been reported as a tumor suppressor gene. Therefore, the aim of this study was to investigate the association between klotho (rs1207568 and rs564481) variants and CRC in Egyptian patients. MATERIALS AND METHODS A case-control study comprising 100 patients with CRC and 100 age- and sex-matched healthy controls was conducted. Genotyping of klotho was performed by polymerase chain reaction with confronting two-pair primers. RESULTS The frequencies of the A allele of rs1207568 and the AC haplotype were significantly higher in patients with CRC than in the controls (p=0.019 and p=0.005, respectively). CONCLUSION We propose that klotho (rs1207568 and rs564481) variants play a significant role in colorectal carcinogenesis and that the klotho protein could be a target for oncotherapy.
Collapse
Affiliation(s)
- Asmaa Kamal
- Department of Clinical - Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed Salama
- Department of Surgical Oncology, National Cancer Institute, Cairo Univeristy, Cairo, Egypt
| | - Amr Kamal
- Department of Surgical Oncology, National Cancer Institute, Cairo Univeristy, Cairo, Egypt
| | - Ahmed Mohsen
- General Surgery Researcher, National Research Centre, Cairo, Egypt
| | - Ibrahem Siam
- Department of Internal Medicine, National Research Centre, Cairo, Egypt
| |
Collapse
|
183
|
Buchanan S, Combet E, Stenvinkel P, Shiels PG. Klotho, Aging, and the Failing Kidney. Front Endocrinol (Lausanne) 2020; 11:560. [PMID: 32982966 PMCID: PMC7481361 DOI: 10.3389/fendo.2020.00560] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022] Open
Abstract
Klotho has been recognized as a gene involved in the aging process in mammals for over 30 years, where it regulates phosphate homeostasis and the activity of members of the fibroblast growth factor (FGF) family. The α-Klotho protein is the receptor for Fibroblast Growth Factor-23 (FGF23), regulating phosphate homeostasis and vitamin D metabolism. Phosphate toxicity is a hallmark of mammalian aging and correlates with diminution of Klotho levels with increasing age. As such, modulation of Klotho activity is an attractive target for therapeutic intervention in the diseasome of aging; in particular for chronic kidney disease (CKD), where Klotho has been implicated directly in the pathophysiology. A range of senotherapeutic strategies have been developed to directly or indirectly influence Klotho expression, with varying degrees of success. These include administration of exogenous Klotho, synthetic and natural Klotho agonists and indirect approaches, via modulation of the foodome and the gut microbiota. All these approaches have significant potential to mitigate loss of physiological function and resilience accompanying old age and to improve outcomes within the diseasome of aging.
Collapse
Affiliation(s)
- Sarah Buchanan
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Emilie Combet
- School of Medicine, Dentistry & Nursing, Human Nutrition, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Peter Stenvinkel
- Division of Renal Medicine M99, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Paul G. Shiels
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
184
|
Bi X, Yang K, Zhang B, Zhao J. The Protective Role of Klotho in CKD-Associated Cardiovascular Disease. KIDNEY DISEASES 2020; 6:395-406. [PMID: 33313060 DOI: 10.1159/000509369] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 06/12/2020] [Indexed: 12/20/2022]
Abstract
Background Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality in advanced CKD. The major pathological changes of CKD-associated CVD are severe vascular media calcification, aberrant cardiac remodeling such as hypertrophy and fibrosis, as well as accelerated atherosclerosis. α-Klotho is proposed as an anti-aging gene, which is primarily expressed in the kidney. Recent studies reveal that α-Klotho deficiency is associated with profound cardiovascular dysfunction. Of note, CKD represents extremely declined α-Klotho levels, hinting that α-Klotho deficiency may be implicated in the pathogenesis of CKD-associated CVD. Summary Based on the pathogenic mechanism of α-Klotho deficiency and decreased Klotho levels in the circulation even early in stage 1 of CKD, α-Klotho serves as a sensitive biomarker for renal insufficiency and also a novel predictor of risk of overall mortality of CVD events in CKD. Meanwhile, loss of Klotho resulted from kidney dysfunction markedly contributes to the progressive development of CKD and CVD. By contrast, prevention of Klotho decline using exogenous supplementation or genetically activated ways by several mechanisms can dramatically mitigate cardiac dysfunction, prevent vascular calcification, and retard the progression of CKD-accelerated atherosclerosis. Key Messages Klotho deficiency is proposed as a novel predictive biomarker as well as a pathogenic contributor to CVD events in CKD. In the future, Klotho may be a crucial potential therapeutic strategy to decrease the burden of CVD comorbidity with CKD in clinics.
Collapse
Affiliation(s)
- Xianjin Bi
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ke Yang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bo Zhang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jinghong Zhao
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
185
|
Podkowińska A, Formanowicz D. Chronic Kidney Disease as Oxidative Stress- and Inflammatory-Mediated Cardiovascular Disease. Antioxidants (Basel) 2020; 9:E752. [PMID: 32823917 PMCID: PMC7463588 DOI: 10.3390/antiox9080752] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Generating reactive oxygen species (ROS) is necessary for both physiology and pathology. An imbalance between endogenous oxidants and antioxidants causes oxidative stress, contributing to vascular dysfunction. The ROS-induced activation of transcription factors and proinflammatory genes increases inflammation. This phenomenon is of crucial importance in patients with chronic kidney disease (CKD), because atherosclerosis is one of the critical factors of their cardiovascular disease (CVD) and increased mortality. The effect of ROS disrupts the excretory function of each section of the nephron. It prevents the maintenance of intra-systemic homeostasis and leads to the accumulation of metabolic products. Renal regulatory mechanisms, such as tubular glomerular feedback, myogenic reflex in the supplying arteriole, and the renin-angiotensin-aldosterone system, are also affected. It makes it impossible for the kidney to compensate for water-electrolyte and acid-base disturbances, which progress further in the mechanism of positive feedback, leading to a further intensification of oxidative stress. As a result, the progression of CKD is observed, with a spectrum of complications such as malnutrition, calcium phosphate abnormalities, atherosclerosis, and anemia. This review aimed to show the role of oxidative stress and inflammation in renal impairment, with a particular emphasis on its influence on the most common disturbances that accompany CKD.
Collapse
Affiliation(s)
| | - Dorota Formanowicz
- Department of Clinical Biochemistry and Laboratory Medicine, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland
| |
Collapse
|
186
|
Xie T, Ye W, Liu J, Zhou L, Song Y. The Emerging Key Role of Klotho in the Hypothalamus-Pituitary-Ovarian Axis. Reprod Sci 2020; 28:322-331. [PMID: 32783104 DOI: 10.1007/s43032-020-00277-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022]
Abstract
The hypothalamus-pituitary-ovary axis is the most important system for regulating female reproductive endocrine function. Its dysfunction would lead to the abnormal secretion of gonadotropin-releasing hormone, follicle-stimulating hormone, or luteinizing hormone, and eventually result in the occurrence of reproductive disease, such as congenital hypogonadotropic hypogonadism, polycystic ovary syndrome, and premature ovarian failure. Recently, an anti-aging gene, Klotho, has gained broad attention in female reproductive diseases. Reports have shown that Klotho is closely correlated to the hypothalamus-pituitary-ovary axis and plays a key role in the development and progression of reproductive diseases. With this issue, we generally review the physiological and pathological role of Klotho in the hypothalamus-pituitary-ovary axis. We also review the underlying mechanisms of Klotho in promoting and preventing female reproductive diseases, which involve the dysfunction of the fibroblast growth factor-Klotho endocrine system, the abnormal signaling regulation of Wnt-β-catenin and insulin-like growth factor-1, the accumulation of oxidative stress, and the inhibition of autophagy, eventually affecting the genesis, development, ovulation, or atresia of follicles. The present review would provide new insights and potential therapeutic target strategies for clinical strategies.
Collapse
Affiliation(s)
- Tingting Xie
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China
| | - Wenting Ye
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China
| | - Jing Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| | - Yali Song
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China.
| |
Collapse
|
187
|
Zhang CY, He FF, Su H, Zhang C, Meng XF. Association between chronic kidney disease and Alzheimer's disease: an update. Metab Brain Dis 2020; 35:883-894. [PMID: 32246323 DOI: 10.1007/s11011-020-00561-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 03/05/2020] [Indexed: 12/11/2022]
Abstract
It has been accepted that kidney function is connected with brain activity. In clinical studies, chronic kidney disease (CKD) patients have been found to be prone to suffering cognitive decline and Alzheimer's disease (AD). The cognitive function of CKD patients may improve after kidney transplantation. All these indicators show a possible link between kidney function and dementia. However, little is known about the mechanism behind the relation of CKD and AD. This review discusses the associations between CKD and AD from the perspective of the pathophysiology of the kidney and complications and/or concomitants of CKD that may lead to cognitive decline in the progression of CKD and AD. Potential preventive and therapeutic strategies for AD are also presented. Further studies are warranted in order to confirm whether the setting of CKD is a possible new determinant for cognitive impairment in AD.
Collapse
Affiliation(s)
- Chun-Yun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fang-Fang He
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hua Su
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xian-Fang Meng
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
188
|
Batlahally S, Franklin A, Damianos A, Huang J, Chen P, Sharma M, Duara J, Keerthy D, Zambrano R, Shehadeh LA, Martinez EC, DeFreitas MJ, Kulandavelu S, Abitbol CL, Freundlich M, Kanashiro-Takeuchi RM, Schmidt A, Benny M, Wu S, Mestan KK, Young KC. Soluble Klotho, a biomarker and therapeutic strategy to reduce bronchopulmonary dysplasia and pulmonary hypertension in preterm infants. Sci Rep 2020; 10:12368. [PMID: 32704023 PMCID: PMC7378054 DOI: 10.1038/s41598-020-69296-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 07/03/2020] [Indexed: 11/09/2022] Open
Abstract
Preterm infants with bronchopulmonary dysplasia (BPD) and pulmonary hypertension (PH) have accelerated lung aging and poor long-term outcomes. Klotho is an antiaging protein that modulates oxidative stress, angiogenesis and fibrosis. Here we test the hypothesis that decreased cord Klotho levels in preterm infants predict increased BPD-PH risk and early Klotho supplementation prevents BPD-like phenotype and PH in rodents exposed to neonatal hyperoxia. In experiment 1, Klotho levels were measured in cord blood of preterm infants who were enrolled in a longitudinal cohort study. In experiment 2, using an experimental BPD-PH model, rat pups exposed to room air or hyperoxia (85% O2) were randomly assigned to receive every other day injections of recombinant Klotho or placebo. The effect of Klotho on lung structure, PH and cardiac function was assessed. As compared to controls, preterm infants with BPD or BPD-PH had decreased cord Klotho levels. Early Klotho supplementation in neonatal hyperoxia-exposed rodents preserved lung alveolar and vascular structure, attenuated PH, reduced pulmonary vascular remodeling and improved cardiac function. Together, these findings have important implications as they suggest that perinatal Klotho deficiency contributes to BPD-PH risk and strategies that preserve Klotho levels, may improve long-term cardiopulmonary outcomes in preterm infants.
Collapse
Affiliation(s)
- Sunil Batlahally
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, RM-345, Miami, FL, USA
| | - Andrew Franklin
- Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andreas Damianos
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, RM-345, Miami, FL, USA
| | - Jian Huang
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, RM-345, Miami, FL, USA
| | - Pingping Chen
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, RM-345, Miami, FL, USA
| | - Mayank Sharma
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, RM-345, Miami, FL, USA
| | - Joanne Duara
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, RM-345, Miami, FL, USA
| | - Divya Keerthy
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, RM-345, Miami, FL, USA
| | - Ronald Zambrano
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, RM-345, Miami, FL, USA
| | - Lina A Shehadeh
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Eliana C Martinez
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Marissa J DeFreitas
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shathiyah Kulandavelu
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Carolyn L Abitbol
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael Freundlich
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rosemeire M Kanashiro-Takeuchi
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Augusto Schmidt
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, RM-345, Miami, FL, USA
| | - Merline Benny
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, RM-345, Miami, FL, USA
| | - Shu Wu
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, RM-345, Miami, FL, USA
| | - Karen K Mestan
- Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karen C Young
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA. .,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, RM-345, Miami, FL, USA. .,The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
189
|
Lloret A, Monllor P, Fuchsberger T, Giraldo E, Perluigi M, Vina J. Increased basal antioxidant levels in RCAN1 - deficient mice lowers oxidative injury after acute paraquat insult. Free Radic Res 2020; 54:442-454. [PMID: 32686528 DOI: 10.1080/10715762.2020.1798002] [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: 10/23/2022]
Abstract
RCAN1 is an inhibitor of the phosphatase calcineurin, which is involved in the regulation of oxidative stress and apoptosis, among other important cell processes. Here we have used RCAN1 deficient mice (RCAN1-/-) to elucidate its role after an acute oxidative insult such as paraquat injection. We have observed that RCAN1-/- mice show less oxidative damage than wildtype (WT) mice after treatment. Under basal conditions, RCAN1-/- animals express more calcineurin, heme oxygenase-1, Nrf2, and catalase compared to WT mice (controls). This may explain the less severe effect of paraquat treatment on RCAN1-/- mice compared to WT. We showed that oxidative stress is involved in the early stages of apoptosis, thus we determined the apoptotic effector BAD and found that decreases in RCAN1-/- mice after treatment with paraquat compared with WT in similar experimental conditions. Our results suggest that RCAN1 may be involved in the balance between oxidant and antioxidant species production in vivo.
Collapse
Affiliation(s)
- Ana Lloret
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES-ISCIII, INCLIVA, Valencia, Spain
| | - Paloma Monllor
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES-ISCIII, INCLIVA, Valencia, Spain
| | - Tanja Fuchsberger
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES-ISCIII, INCLIVA, Valencia, Spain
| | - Esther Giraldo
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES-ISCIII, INCLIVA, Valencia, Spain.,Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain.,The Principe Felipe Research Center, Valencia, Spain
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Jose Vina
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES-ISCIII, INCLIVA, Valencia, Spain
| |
Collapse
|
190
|
Wu CM, Zheng L, Wang Q, Hu YW. The emerging role of cell senescence in atherosclerosis. Clin Chem Lab Med 2020; 59:27-38. [PMID: 32692694 DOI: 10.1515/cclm-2020-0601] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 06/15/2020] [Indexed: 12/15/2022]
Abstract
Cell senescence is a fundamental mechanism of aging and appears to play vital roles in the onset and prognosis of cardiovascular disease, fibrotic pulmonary disease, liver disease and tumor. Moreover, an increasing body of evidence shows that cell senescence plays an indispensable role in the formation and development of atherosclerosis. Multiple senescent cell types are associated with atherosclerosis, senescent human vascular endothelial cells participated in atherosclerosis via regulating the level of endothelin-1 (ET-1), nitric oxide (NO), angiotensin II and monocyte chemoattractant protein-1 (MCP-1), senescent human vascular smooth muscle cells-mediated plaque instability and vascular calcification via regulating the expression level of BMP-2, OPN, Runx-2 and inflammatory molecules, and senescent macrophages impaired cholesterol efflux and promoted the development of senescent-related cardiovascular diseases. This review summarizes the characteristics of cell senescence and updates the molecular mechanisms underlying cell senescence. Moreover, we also discuss the recent advances on the molecular mechanisms that can potentially regulate the development and progression of atherosclerosis.
Collapse
Affiliation(s)
- Chang-Meng Wu
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Lei Zheng
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Qian Wang
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Yan-Wei Hu
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China.,Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou, P. R. China
| |
Collapse
|
191
|
Kitada M, Xu J, Ogura Y, Monno I, Koya D. Manganese Superoxide Dismutase Dysfunction and the Pathogenesis of Kidney Disease. Front Physiol 2020; 11:755. [PMID: 32760286 PMCID: PMC7373076 DOI: 10.3389/fphys.2020.00755] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
The mitochondria are a major source of reactive oxygen species (ROS). Superoxide anion (O2•–) is produced by the process of oxidative phosphorylation associated with glucose, amino acid, and fatty acid metabolism, resulting in the production of adenosine triphosphate (ATP) in the mitochondria. Excess production of reactive oxidants in the mitochondria, including O2•–, and its by-product, peroxynitrite (ONOO–), which is generated by a reaction between O2•– with nitric oxide (NO•), alters cellular function via oxidative modification of proteins, lipids, and nucleic acids. Mitochondria maintain an antioxidant enzyme system that eliminates excess ROS; manganese superoxide dismutase (Mn-SOD) is one of the major components of this system, as it catalyzes the first step involved in scavenging ROS. Reduced expression and/or the activity of Mn-SOD results in diminished mitochondrial antioxidant capacity; this can impair the overall health of the cell by altering mitochondrial function and may lead to the development and progression of kidney disease. Targeted therapeutic agents may protect mitochondrial proteins, including Mn-SOD against oxidative stress-induced dysfunction, and this may consequently lead to the protection of renal function. Here, we describe the biological function and regulation of Mn-SOD and review the significance of mitochondrial oxidative stress concerning the pathogenesis of kidney diseases, including chronic kidney disease (CKD) and acute kidney injury (AKI), with a focus on Mn-SOD dysfunction.
Collapse
Affiliation(s)
- Munehiro Kitada
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan.,Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| | - Jing Xu
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Yoshio Ogura
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Itaru Monno
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Japan.,Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
192
|
Sorrenti V, Davinelli S, Scapagnini G, Willcox BJ, Allsopp RC, Willcox DC. Astaxanthin as a Putative Geroprotector: Molecular Basis and Focus on Brain Aging. Mar Drugs 2020; 18:md18070351. [PMID: 32635607 PMCID: PMC7401246 DOI: 10.3390/md18070351] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/26/2020] [Accepted: 07/01/2020] [Indexed: 12/16/2022] Open
Abstract
In recent years, the scientific interest in natural compounds with geroprotective activities has grown exponentially. Among the various naturally derived molecules, astaxanthin (ASX) represents a highly promising candidate geroprotector. By virtue of the central polyene chain, ASX acts as a scavenger of free radicals in the internal membrane layer and simultaneously controls oxidation on the membrane surface. Moreover, several studies have highlighted ASX’s ability to modulate numerous biological mechanisms at the cellular level, including the modulation of transcription factors and genes directly linked to longevity-related pathways. One of the main relevant evolutionarily-conserved transcription factors modulated by astaxanthin is the forkhead box O3 gene (FOXO3), which has been recognized as a critical controller of cell fate and function. Moreover, FOXO3 is one of only two genes shown to robustly affect human longevity. Due to its tropism in the brain, ASX has recently been studied as a putative neuroprotective molecule capable of delaying or preventing brain aging in different experimental models of brain damage or neurodegenerative diseases. Astaxanthin has been observed to slow down brain aging by increasing brain-derived neurotrophic factor (BDNF) levels in the brain, attenuating oxidative damage to lipids, protein, and DNA and protecting mitochondrial functions. Emerging data now suggest that ASX can modulate Nrf2, FOXO3, Sirt1, and Klotho proteins that are linked to longevity. Together, these mechanisms provide support for a role of ASX as a potential geroneuroprotector.
Collapse
Affiliation(s)
- Vincenzo Sorrenti
- Department of Pharmaceutical Pharmacological Sciences, University of Padova, 35131 Padova, Italy
- Bendessere™ Study Center, 35131 Padova, Italy
- Correspondence:
| | - Sergio Davinelli
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Via de Sanctis s.n.c, 86100 Campobasso, Italy; (S.D.); (G.S.)
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Via de Sanctis s.n.c, 86100 Campobasso, Italy; (S.D.); (G.S.)
| | - Bradley J. Willcox
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96817, USA; (B.J.W.); (D.C.W.)
- Department of Research, Kuakini Medical Center, Honolulu, HI 96817, USA
| | - Richard C. Allsopp
- Department of Anatomy and Reproductive Biology, Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA;
| | - Donald C. Willcox
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96817, USA; (B.J.W.); (D.C.W.)
- Department of Research, Kuakini Medical Center, Honolulu, HI 96817, USA
- Department of Human Welfare, Okinawa International University, Ginowan 901-2701, Japan
| |
Collapse
|
193
|
Wolf EJ, Logue MW, Zhao X, Daskalakis NP, Morrison FG, Escarfulleri S, Stone A, Schichman SA, McGlinchey RE, Milberg WP, Chen C, Abraham CR, Miller MW. PTSD and the klotho longevity gene: Evaluation of longitudinal effects on inflammation via DNA methylation. Psychoneuroendocrinology 2020; 117:104656. [PMID: 32438247 PMCID: PMC7293549 DOI: 10.1016/j.psyneuen.2020.104656] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/04/2020] [Accepted: 03/18/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Longevity gene klotho (KL) is associated with age-related phenotypes including lifespan, cardiometabolic disorders, cognition, and brain morphology, in part, by conferring protection against inflammation. We hypothesized that the KL/inflammation association might be altered in the presence of psychiatric stress and operate via epigenetic pathways. We examined KL polymorphisms, and their interaction with posttraumatic stress disorder (PTSD) symptoms, in association with KL DNA methylation in blood. We further examined KL DNA methylation as a predictor of longitudinal changes in a peripheral biomarker of inflammation (C-reactive protein; CRP). METHODS The sample comprised 309 white non-Hispanic military veterans (93.5 % male; mean age: 32 years, range: 19-65; 30 % PTSD per structured diagnostic interview); 111 were reassessed approximately two years later. RESULTS Analyses revealed a methylation quantitative trait locus at rs9527025 (C370S, previously implicated in numerous studies of aging) in association with a Cytosine-phosphate-Guanine site (cg00129557; B = -.65, p = 1.29 X 10-20), located within a DNase hypersensitivity site in the body of KL. There was also a rs9527025 x PTSD severity interaction (B = .004, p = .035) on methylation at this locus such that the minor allele was associated with reduced cg00129557 methylation in individuals with few or no PTSD symptoms while this effect was attenuated in those with elevated levels of PTSD. Path models revealed that methylation at cg00129557 was inversely associated with CRP over time (B = -.14, p = .005), controlling for baseline CRP. There was also an indirect effect of rs9527025 X PTSD on subsequent CRP via cg00129557 methylation (indirect B = -.002, p = .033). CONCLUSIONS Results contribute to our understanding of the epigenetic correlates of inflammation in PTSD and suggest that KL methylation may be a mechanism by which KL genotype confers risk vs. resilience to accelerated aging in those experiencing traumatic stress.
Collapse
Affiliation(s)
- Erika J. Wolf
- National Center for PTSD at VA Boston Healthcare System,Department of Psychiatry, Boston University School of Medicine
| | - Mark W. Logue
- National Center for PTSD at VA Boston Healthcare System,Department of Psychiatry, Boston University School of Medicine,Biomedical Genetics, Boston University School of Medicine
| | - Xiang Zhao
- National Center for PTSD at VA Boston Healthcare System,Department of Psychiatry, Boston University School of Medicine
| | | | - Filomene G. Morrison
- National Center for PTSD at VA Boston Healthcare System,Department of Psychiatry, Boston University School of Medicine
| | | | - Annjanette Stone
- Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System
| | - Steven A. Schichman
- Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System
| | - Regina E. McGlinchey
- Geriatric Research Educational and Clinical Center and Translational Research Center for TBI and Stress Disorders, VA Boston Healthcare System,Department of Psychiatry, Harvard Medical School
| | - William P. Milberg
- Geriatric Research Educational and Clinical Center and Translational Research Center for TBI and Stress Disorders, VA Boston Healthcare System,Department of Psychiatry, Harvard Medical School
| | - Cidi Chen
- Department of Biochemistry, Boston University School of Medicine
| | - Carmela R. Abraham
- Department of Biochemistry, Boston University School of Medicine,Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine
| | - Mark W. Miller
- National Center for PTSD at VA Boston Healthcare System,Department of Psychiatry, Boston University School of Medicine
| |
Collapse
|
194
|
Bao JF, Hu PP, She QY, Li A. A Land of Controversy: Fibroblast Growth Factor-23 and Uremic Cardiac Hypertrophy. J Am Soc Nephrol 2020; 31:1423-1434. [PMID: 32527977 PMCID: PMC7351013 DOI: 10.1681/asn.2020010081] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiac hypertrophy is a common feature in patients with CKD. Recent studies revealed that two phosphate regulators, fibroblast growth factor-23 and α-Klotho, are highly involved in the pathophysiologic process of CKD-induced cardiac hypertrophy. With decreasing renal function, elevated fibroblast growth factor-23 and decreased α-Klotho may contribute to cardiac hypertrophy by targeting the heart directly or by inducing systemic changes, such as vascular injury, hemodynamic disorders, and inflammation. However, several studies have demonstrated that disturbances in the fibroblast growth factor-23/α-Klotho axis do not lead to cardiac hypertrophy. In this review, we describe the cardiac effects of the fibroblast growth factor-23/α-Klotho axis and summarize recent progress in this field. In addition, we present not only the main controversies in this field but also provide possible directions to resolve these disputes.
Collapse
Affiliation(s)
- Jing-Fu Bao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pan-Pan Hu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qin-Ying She
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Aiqing Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
195
|
Miranda Pérez AA, Gutiérrez Pérez ME, Urraza Robledo AI, Delgadillo Guzmán D, Ruíz Flores P, López Márquez FC. Klotho-HIV and Oxidative Stress: The Role of Klotho in Cardiovascular Disease Under HIV Infection-A Review. DNA Cell Biol 2020; 39:1478-1485. [PMID: 32584609 DOI: 10.1089/dna.2020.5444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Combined antiretroviral therapy has improved quality and life expectancy of people living with human immunodeficiency virus (HIV). However, this therapy increases oxidative stress (OS), which in turn causes alterations in lipid and carbon metabolism, kidney disease, liver cirrhosis, and increased risk of cardiovascular disease. The Klotho gene has been implicated in cardiovascular risk increase. Klotho protein expression at X level decreases the risk of heart disease. HIV-positive people usually present low plasma levels of Klotho; thus, contributing to some extent to an increase in cardiovascular risk for these types of patients, mostly by favoring atherosclerosis. Therefore, our aim is to provide an overview of the effect of OS on Klotho protein and its consequent cardiometabolic alterations in HIV-positive patients on antiretroviral therapy.
Collapse
Affiliation(s)
- Alberto Alejandro Miranda Pérez
- Department of Molecular Immunobiology, Biomedical Research Center, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | - María Elena Gutiérrez Pérez
- Department of Molecular Immunobiology, Biomedical Research Center, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | | | - Dealmy Delgadillo Guzmán
- Department of Pharmacology, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | - Pablo Ruíz Flores
- Department of Genetics, Center for Biomedical Research Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | - Francisco Carlos López Márquez
- Department of Molecular Immunobiology, Biomedical Research Center, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| |
Collapse
|
196
|
Klotho and fibroblast growth factors 19 and 21 serum concentrations in children and adolescents with normal body weight and obesity and their associations with metabolic parameters. BMC Pediatr 2020; 20:294. [PMID: 32546231 PMCID: PMC7296965 DOI: 10.1186/s12887-020-02199-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Fibroblast growth factor 19 (FGF19), fibroblast growth factor 21 (FGF21) and Klotho are regulators of energy homeostasis. However, in the pediatric population, the relationships between obesity, metabolic disorders and the aforementioned factors have not been clearly investigated. We analyzed the role of FGF19, FGF21 and Klotho protein in children with normal body weight as well as in overweight and obese subjects and explored their associations with insulin resistance (IR) and metabolic syndrome (MS) and its components. METHODS This was a cross-sectional study conducted in a group of hospitalized children and adolescents. Laboratory investigations included serum analysis of FGF19, FGF21, and Klotho with ELISA kits as well as the analysis of the lipid profile and ALT serum concentrations. Moreover, each subject underwent an oral glucose tolerance test (OGTT) with fasting insulinemia measurement to detect glucose tolerance abnormalities and calculate the Homeostatic Model Assessment of Insulin Resistance (HOMA-IR) index. Furthermore, the clinical analysis included blood pressure measurement, body fat percentage estimation and assessment of the prevalence of MS and its components. RESULTS The study was conducted with 174 children/adolescents aged 6-17 years with normal body weight (N = 48), obesity (N = 92) and overweight (N = 34). Klotho concentration was significantly higher in the obese children [median 168.6 pg/ml (90.2 to 375.9)]) than in the overweight [131.3 pg/ml (78.0 to 313.0)] and normal-body-weight subjects [116.6 pg/ml (38.5 to 163.9)] (p = 0.0334) and was also significantly higher in insulin-resistant children than in insulin-sensitive children [185.3 pg/ml (102.1 to 398.2) vs 132.6 pg/ml (63.9 to 275.6), p = 0.0283]. FGF21 was elevated in patients with MS compared to the FGF21 levels in other subjects [136.2 pg/ml (86.5 to 239.9) vs 82.6 pg/ml (41.8 to 152.4), p = 0.0286]. The multivariable model showed that FGF19 was an independent predictor of IR after adjusting for pubertal stage and BMI Z-score. CONCLUSIONS Klotho levels were associated with body weight status in children and adolescents. Moreover, Klotho, FGF19 and FGF21 concentrations correlated with IR status and/or components of MS.
Collapse
|
197
|
Xiong JW, Zhan JQ, Luo T, Chen HB, Wan QG, Wang Y, Wei B, Yang YJ. Increased Plasma Level of Longevity Protein Klotho as a Potential Indicator of Cognitive Function Preservation in Patients With Schizophrenia. Front Neurosci 2020; 14:610. [PMID: 32612508 PMCID: PMC7308714 DOI: 10.3389/fnins.2020.00610] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/18/2020] [Indexed: 01/02/2023] Open
Abstract
Cognitive impairments are a core feature of schizophrenia. Klotho is an anti-aging protein with demonstrated cognitive-enhancing effects on the brain. The purpose of this study was to investigate the differences in levels of plasma klotho between patients with schizophrenia and healthy controls, as well as the relationship between klotho level and cognitive function in patients. Forty patients with schizophrenia and 40 gender- and age-matched healthy individuals were recruited. Positive and Negative Syndrome Scale (PANSS) was used to assess the psychopathology of patients. A neuropsychological battery was performed to evaluate the cognitive function of participants. Plasma klotho was measured using enzyme-linked immunosorbent assay. We show that patients with schizophrenia performed worse in the neurocognitive tests than the healthy controls. The levels of plasma klotho were significantly higher in schizophrenia patients than in healthy controls (p < 0.001). In patients, plasma klotho levels were positively correlated with cognitive function with regard to attention (p = 0.010), working memory (p < 0.001), verbal memory (p = 0.044), executive function (p < 0.001), and composite cognitive score (p < 0.001). Stepwise linear regression analysis shows that executive function had the highest correlation with plasma klotho levels (β = 0.896, t = 8.290, p < 0.001). Collectively, these results indicate that anti-aging protein klotho may be implicated in the pathogenesis of schizophrenia, and increased klotho may act as a compensatory factor for the preservation of cognitive function in schizophrenia. Further studies are needed to investigate the dynamic changes of klotho and the mechanisms by which klotho modulates cognition in schizophrenia.
Collapse
Affiliation(s)
- Jian-wen Xiong
- Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| | - Jin-qiong Zhan
- Biological Psychiatry Laboratory, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| | - Tao Luo
- Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| | - Hai-bo Chen
- Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| | - Qi-gen Wan
- Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| | - Yan Wang
- Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| | - Bo Wei
- Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
- Biological Psychiatry Laboratory, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| | - Yuan-jian Yang
- Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
- Biological Psychiatry Laboratory, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
198
|
Sodium butyrate protects against oxidative stress in human nucleus pulposus cells via elevating PPARγ-regulated Klotho expression. Int Immunopharmacol 2020; 85:106657. [PMID: 32554208 DOI: 10.1016/j.intimp.2020.106657] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/15/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023]
Abstract
We investigated the involvement of klotho in the inhibition of oxidative stress by sodium butyrate (NaB) in human nucleus pulposus cells (NPCs). NPCs were pretreated with different concentrations of NaB for 2 h before stimulation with tert-butyl hydroperoxide (TBHP). NaB alleviated TBHP-induced oxidative injury in the NPCs, as evident by the reduced accumulation of mitochondrial superoxide, intracellular reactive oxygen species, and malondialdehyde, and increased activities of superoxide dismutase and glutathione peroxidase. Flow cytometry and western blotting showed that TBHP-induced apoptosis of NPCs was inhibited by NaB. NaB also reduced the TBHP-induced release of proteases that degrade the extracellular matrix, including matrix metalloproteinases 3 and 13, and ADAMTS-4 (a disintegrin and metalloproteinase with thrombospondin motifs 4). Intriguingly, NaB significantly reversed TBHP-induced klotho suppression. However, the protective effects of NaB on NPCs were abolished by klotho-specific small interfering RNA (siRNA). TBHP stimulation had no obvious effects on total or nuclear expression of peroxisome proliferator-activated receptor γ (PPARγ), but significantly reduced PPARγ acetylation and transcriptional activity, which were restored by NaB. TBHP stimulation also promoted the nuclear translocation of histone deacetylase 3 (HDAC3) and enhanced the association between HDAC3 and PPARγ in the nucleus, but this interaction was substantially disrupted by NaB. siRNA-induced HDAC3 knockdown significantly increased PPARγ acetylation and transactivation, reversing the TBHP-induced suppression of klotho. Therefore, NaB alleviates TBHP-induced oxidative stress in human NPCs by elevating PPARγ-regulated klotho expression. HDAC3 may be a critical HDAC subtype that mediates the regulation of PPARγ activity by NaB under oxidative stress.
Collapse
|
199
|
Guo X, Kassab GS. Increased Serum Klotho With Age-Related Aortic Stiffness and Peripheral Vascular Resistance in Young and Middle-Aged Swine. Front Physiol 2020; 11:591. [PMID: 32581850 PMCID: PMC7297143 DOI: 10.3389/fphys.2020.00591] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022] Open
Abstract
The anti-aging function of Klotho gene has been implicated in age-related diseases. The physiological importance of Klotho in the progression of arterial stiffness with aging, however, remains unclear. The goal of this study is to determine the correlation of circulating Klotho with early age-related aortic stiffening and peripheral hemodynamics. We measured serum Klotho levels in a group of pigs with age ranges of 1.5-9 years and investigated the relationship between Klotho levels and biomarkers of aortic stiffening with aging, including aortic pulse wave velocity (PWV), augmentation index (AIx), and pulse pressure (PP). The effects of aortic stiffening on peripheral vascular resistance, compliance, and function were also evaluated. We found that increased aortic stiffness occurred at middle age (>5 years old), as evidenced by an increase in PWV and AIx (p < 0.001), but with no changes in blood pressure and PP. With advancing age, increased femoral vascular resistance positively correlated with aortic PWV and AIx (p < 0.01). No significant difference in endothelium function and arterial compliance for femoral and small peripheral arteries was observed between young and middle-aged groups. The serum Klotho levels were lower in young and higher in middle-aged pigs (p < 0.001), and a positive correlation was found between Klotho and aortic PWV, AIx, and femoral vascular resistance (p < 0.01). Our findings suggest that early-aged aortic stiffening has adverse effect on peripheral hemodynamics, independent of blood pressure levels. Elevated Klotho secretion was associated with increased aortic stiffness and peripheral vascular resistance with aging.
Collapse
Affiliation(s)
| | - Ghassan S. Kassab
- California Medical Innovations Institute, San Diego, CA, United States
| |
Collapse
|
200
|
Liberale L, Camici GG. The Role of Vascular Aging in Atherosclerotic Plaque Development and Vulnerability. Curr Pharm Des 2020; 25:3098-3111. [PMID: 31470777 DOI: 10.2174/1381612825666190830175424] [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: 07/19/2019] [Accepted: 08/24/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND The ongoing demographical shift is leading to an unprecedented aging of the population. As a consequence, the prevalence of age-related diseases, such as atherosclerosis and its thrombotic complications is set to increase in the near future. Endothelial dysfunction and vascular stiffening characterize arterial aging and set the stage for the development of cardiovascular diseases. Atherosclerotic plaques evolve over time, the extent to which these changes might affect their stability and predispose to sudden complications remains to be determined. Recent advances in imaging technology will allow for longitudinal prospective studies following the progression of plaque burden aimed at better characterizing changes over time associated with plaque stability or rupture. Oxidative stress and inflammation, firmly established driving forces of age-related CV dysfunction, also play an important role in atherosclerotic plaque destabilization and rupture. Several genes involved in lifespan determination are known regulator of redox cellular balance and pre-clinical evidence underlines their pathophysiological roles in age-related cardiovascular dysfunction and atherosclerosis. OBJECTIVE The aim of this narrative review is to examine the impact of aging on arterial function and atherosclerotic plaque development. Furthermore, we report how molecular mechanisms of vascular aging might regulate age-related plaque modifications and how this may help to identify novel therapeutic targets to attenuate the increased risk of CV disease in elderly people.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, CH-8952 Schlieren, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, CH-8952 Schlieren, Switzerland.,University Heart Center, University Hospital Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland.,Department of Research and Education, University Hospital Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| |
Collapse
|