1
|
Yeom E, Mun H, Lim J, Chun YL, Min KW, Lambert J, Cowart LA, Pierce JS, Ogretmen B, Cho JH, Chang JH, Buchan JR, Pitt J, Kaeberlein M, Kang SU, Kwon ES, Ko S, Choi KM, Lee YS, Ha YS, Kim SJ, Lee KP, Kim HS, Yang SY, Shin CH, Yoon JH, Lee KS. Phosphorylation of an RNA-Binding Protein Rck/Me31b by Hippo Is Essential for Adipose Tissue Aging. Aging Cell 2025:e70022. [PMID: 40070010 DOI: 10.1111/acel.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/20/2025] [Accepted: 02/10/2025] [Indexed: 05/15/2025] Open
Abstract
The metazoan lifespan is determined in part by a complex signaling network that regulates energy metabolism and stress responses. Key signaling hubs in this network include insulin/IGF-1, AMPK, mTOR, and sirtuins. The Hippo/Mammalian Ste20-like Kinase1 (MST1) pathway has been reported to maintain lifespan in Caenorhabditis elegans, but its role has not been studied in higher metazoans. In this study, we report that overexpression of Hpo, the MST1 homolog in Drosophila melanogaster, decreased lifespan with concomitant changes in lipid metabolism and aging-associated gene expression, while RNAi Hpo depletion increased lifespan. These effects were mediated primarily by Hpo-induced transcriptional activation of the RNA-binding protein maternal expression at 31B (Me31b)/RCK, resulting in stabilization of mRNA-encoding a lipolytic hormone, Akh. In mouse adipocytes, Hpo/Mst1 mediated adipocyte differentiation, phosphorylation of RNA-binding proteins such as Rck, decapping MRNA 2 (Dcp2), enhancer Of MRNA decapping 3 (Edc3), nucleolin (NCL), and glucagon mRNA stability by interacting with Rck. Decreased lifespan in Hpo-overexpressing Drosophila lines required expression of Me31b, but not DCP2, which was potentially mediated by recovering expression of lipid metabolic genes and formation of lipid droplets. Taken together, our findings suggest that Hpo/Mst1 plays a conserved role in longevity by regulating adipogenesis and fatty acid metabolism.
Collapse
Affiliation(s)
- Eunbyul Yeom
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu, Korea
- Neurophysiology and Metabolism Research Group, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Hyejin Mun
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Oncology Science, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Jinhwan Lim
- Department of Environmental and Occupational Heatlh, University of California, Irvine, California, USA
- Translational Gerontology Branch, National Institute of Aging Intramural Research Program, Baltimore, Maryland, USA
| | - Yoo Lim Chun
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kyung-Won Min
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Biology, College of Natural Sciences, Gangneung-Wonju National University, Gangneung, South Korea
| | - Johana Lambert
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
- Hunter Holmes McGuire Veteran's Affairs Medical Center, Richmond, Virginia, USA
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
- Hunter Holmes McGuire Veteran's Affairs Medical Center, Richmond, Virginia, USA
| | - Jason S Pierce
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jung-Hyun Cho
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jeong Ho Chang
- Department of Biology Education, Kyungpook National University, Daegu, Republic of Korea
| | - J Ross Buchan
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, USA
| | - Jason Pitt
- Department of Laboratory Medicine and Pathology, University of Washington, Washington, DC, USA
| | - Matt Kaeberlein
- Department of Laboratory Medicine and Pathology, University of Washington, Washington, DC, USA
| | - Sung-Ung Kang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eun-Soo Kwon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Seungbeom Ko
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kyoung-Min Choi
- Department of Oncology Science, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Yong Sun Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Yoon-Su Ha
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Seung-Jin Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Kwang-Pyo Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Hyo-Sung Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu, Korea
| | - Seo Young Yang
- Department of Biology Education, Kyungpook National University, Daegu, Republic of Korea
| | - Chang Hoon Shin
- Department of Oncology Science, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Je-Hyun Yoon
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Oncology Science, University of Oklahoma, Oklahoma City, Oklahoma, USA
- Department of Pathology, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Kyu-Sun Lee
- Neurophysiology and Metabolism Research Group, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
2
|
Coleman AE, Creevy KE, Anderson R, Reed MJ, Fajt VR, Aicher KM, Atiee G, Barnett BG, Baumwart RD, Boudreau B, Cunningham SM, Dunbar MD, Ditzler B, Ferguson AM, Forsyth KK, Gambino AN, Gordon SG, Hammond HK, Holland SN, Iannaccone MK, Illing K, Kadotani S, Knowles SA, MacLean EL, Maran BA, Markovic LE, McGrath S, Melvin RL, Mueller MS, Nelson OL, Olby NJ, Pancotto TE, Parsley E, Potter BM, Prescott JO, Saunders AB, Sawyer HM, Scansen BA, Schmid SM, Smith CC, Tjostheim SS, Tolbert MK, Tropf MA, Visser LC, Ward JL, Wesselowski SR, Windsor RC, Yang VK, Ruple A, Promislow DEL, Kaeberlein M. Test of Rapamycin in Aging Dogs (TRIAD): study design and rationale for a prospective, parallel-group, double-masked, randomized, placebo-controlled, multicenter trial of rapamycin in healthy middle-aged dogs from the Dog Aging Project. GeroScience 2025:10.1007/s11357-024-01484-7. [PMID: 39951177 DOI: 10.1007/s11357-024-01484-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/14/2024] [Indexed: 02/25/2025] Open
Abstract
Companion dogs are a powerful model for aging research given their morphologic and genetic variability, risk for age-related disease, and habitation of the human environment. In addition, the shorter life expectancy of dogs compared to human beings provides a unique opportunity for an accelerated timeline to test interventions that might extend healthy lifespan. The Test of Rapamycin In Aging Dogs (TRIAD) randomized clinical trial is a parallel-group, double-masked, randomized, placebo-controlled, multicenter trial that will test the ability of rapamycin to prolong lifespan and improve several healthspan metrics in healthy, middle-aged dogs recruited from Dog Aging Project participants. Here, we describe the rationale, design, and goals of the TRIAD randomized clinical trial, the first rigorous test of a pharmacologic intervention against biological aging with lifespan and healthspan metrics as endpoints to be performed outside of the laboratory in any species.
Collapse
Affiliation(s)
- Amanda E Coleman
- Department of Small Animal Medicine and Surgery, University of Georgia, Athens, GA, USA.
| | - Kate E Creevy
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Rozalyn Anderson
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- GRECC William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - May J Reed
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Virginia R Fajt
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Kathleen M Aicher
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Genna Atiee
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Brian G Barnett
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Ryan D Baumwart
- Department of Veterinary Clinical Sciences, Washington State University, Pullman, WA, USA
| | - Beth Boudreau
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | | | - Matthew D Dunbar
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, USA
| | - Bobbie Ditzler
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Anna M Ferguson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kiersten K Forsyth
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Anya N Gambino
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Sonya G Gordon
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Hillary K Hammond
- Department of Small Animal Medicine and Surgery, University of Georgia, Athens, GA, USA
| | - Sydney N Holland
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Mary K Iannaccone
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Kate Illing
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Saki Kadotani
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana, IL, USA
| | - Shelby A Knowles
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Evan L MacLean
- College of Veterinary Medicine, University of Arizona, Tucson, AZ, USA
| | | | - Lauren E Markovic
- Department of Small Animal Medicine and Surgery, University of Georgia, Athens, GA, USA
| | - Stephanie McGrath
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Rachel L Melvin
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | - O Lynne Nelson
- Department of Veterinary Clinical Sciences, Washington State University, Pullman, WA, USA
| | - Natasha J Olby
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, USA
| | | | - Elizabeth Parsley
- Department of Clinical Sciences, Tufts University, North Grafton, MA, USA
| | - Brianna M Potter
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Jena O Prescott
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Ashley B Saunders
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | | | - Brian A Scansen
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Sarah M Schmid
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | | | - Sonja S Tjostheim
- Department of Medical Sciences, University of Wisconsin, Madison, WI, USA
| | - M Katherine Tolbert
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | - Melissa A Tropf
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, USA
| | - Lance C Visser
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Jessica L Ward
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, USA
| | - Sonya R Wesselowski
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | | | - Vicky K Yang
- Department of Clinical Sciences, Tufts University, North Grafton, MA, USA
| | - Audrey Ruple
- Department of Population and Health Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Daniel E L Promislow
- Jean Mayer USDA Human Nutrition Research Center On Aging, Tufts University, Boston, MA, USA
| | - Matt Kaeberlein
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Optispan, Inc, Seattle, WA, USA
| |
Collapse
|
3
|
Thomas SD, Jayaprakash P, Marwan NZHJ, Aziz EABA, Kuder K, Łażewska D, Kieć-Kononowicz K, Sadek B. Alleviation of Autophagic Deficits and Neuroinflammation by Histamine H3 Receptor Antagonist E159 Ameliorates Autism-Related Behaviors in BTBR Mice. Pharmaceuticals (Basel) 2024; 17:1293. [PMID: 39458934 PMCID: PMC11510413 DOI: 10.3390/ph17101293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Autism spectrum disorder (ASD) is a neurodevelopmental condition marked by social interaction difficulties, repetitive behaviors, and immune dysregulation with elevated pro-inflammatory markers. Autophagic deficiency also contributes to social behavior deficits in ASD. Histamine H3 receptor (H3R) antagonism is a potential treatment strategy for brain disorders with features overlapping ASD, such as schizophrenia and Alzheimer's disease. METHODS This study investigated the effects of sub-chronic systemic treatment with the H3R antagonist E159 on social deficits, repetitive behaviors, neuroinflammation, and autophagic disruption in male BTBR mice. RESULTS E159 (2.5, 5, and 10 mg/kg, i.p.) improved stereotypic repetitive behavior by reducing self-grooming time and enhancing spontaneous alternation in addition to attenuating social deficits. It also decreased pro-inflammatory cytokines in the cerebellum and hippocampus of treated BTBR mice. In BTBR mice, reduced expression of autophagy-related proteins LC3A/B and Beclin 1 was observed, which was elevated following treatment with E159, attenuating the disruption in autophagy. The co-administration with the H3R agonist MHA (10 mg/kg, i.p.) reversed these effects, highlighting the role of histaminergic neurotransmission in observed behavioral improvements. CONCLUSIONS These preliminary findings suggest the therapeutic potential of H3R antagonists in targeting neuroinflammation and autophagic disruption to improve ASD-like behaviors.
Collapse
Affiliation(s)
- Shilu Deepa Thomas
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Petrilla Jayaprakash
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Nurfirzana Z. H. J. Marwan
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Ezzatul A. B. A. Aziz
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Kamil Kuder
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna Str. 9, 30-688 Kraków, Poland; (K.K.); (D.Ł.); (K.K.-K.)
| | - Dorota Łażewska
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna Str. 9, 30-688 Kraków, Poland; (K.K.); (D.Ł.); (K.K.-K.)
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna Str. 9, 30-688 Kraków, Poland; (K.K.); (D.Ł.); (K.K.-K.)
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (S.D.T.); (P.J.); (N.Z.H.J.M.); (E.A.B.A.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
4
|
Kaeberlein TL, Green AS, Haddad G, Hudson J, Isman A, Nyquist A, Rosen BS, Suh Y, Zalzala S, Zhang X, Blagosklonny MV, An JY, Kaeberlein M. Evaluation of off-label rapamycin use to promote healthspan in 333 adults. GeroScience 2023; 45:2757-2768. [PMID: 37191826 PMCID: PMC10187519 DOI: 10.1007/s11357-023-00818-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/02/2023] [Indexed: 05/17/2023] Open
Abstract
Rapamycin (sirolimus) is an FDA-approved drug with immune-modulating and growth-inhibitory properties. Preclinical studies have shown that rapamycin extends lifespan and healthspan metrics in yeast, invertebrates, and rodents. Several physicians are now prescribing rapamycin off-label as a preventative therapy to maintain healthspan. Thus far, however, there is limited data available on side effects or efficacy associated with use of rapamycin in this context. To begin to address this gap in knowledge, we collected data from 333 adults with a history of off-label use of rapamycin by survey. Similar data were also collected from 172 adults who had never used rapamycin. Here, we describe the general characteristics of a patient cohort using off-label rapamycin and present initial evidence that rapamycin can be used safely in adults of normal health status.
Collapse
Affiliation(s)
- Tammi L Kaeberlein
- Department of Oral Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | | | | | - Johnny Hudson
- Department of Oral Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | | | | | | | - Yousin Suh
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | | | - Xingyu Zhang
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, 15219, USA
| | | | - Jonathan Y An
- Department of Oral Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Matt Kaeberlein
- Optispan Geroscience, Seattle, WA, 98168, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
5
|
Barnett BG, Wesselowski SR, Gordon SG, Saunders AB, Promislow DEL, Schwartz SM, Chou L, Evans JB, Kaeberlein M, Creevy KE. A masked, placebo-controlled, randomized clinical trial evaluating safety and the effect on cardiac function of low-dose rapamycin in 17 healthy client-owned dogs. Front Vet Sci 2023; 10:1168711. [PMID: 37275618 PMCID: PMC10233048 DOI: 10.3389/fvets.2023.1168711] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 05/03/2023] [Indexed: 06/07/2023] Open
Abstract
Introduction Geroscience studies of low-dose rapamycin in laboratory species have identified numerous benefits, including reversing age-related cardiac dysfunction. Cardiovascular benefits have been observed in dogs with 10 weeks of treatment, raising questions about possible benefits and adverse effects of long-term use of low-dose rapamycin. The objectives of this study were to assess the impact of 6 months of low-dose rapamycin on echocardiographic indices of cardiac function in healthy dogs and to document the occurrence of adverse events. Methods Seventeen client-owned dogs aged 6-10 years, weighing 18-36 kg, and without significant systemic disease were included in a prospective, randomized, placebo-controlled, masked clinical trial. Low-dose rapamycin (0.025 mg/kg) or placebo was administered three times per week for 6 months. Baseline, 6-month, and 12-month evaluation included physical examination, cardiology examination, and clinicopathology. Three-month evaluation included physical examination and clinicopathology. Owners completed online questionnaires every 2 weeks. Results There were no statistically significant differences in echocardiographic parameters between rapamycin and placebo groups at 6 or 12 months. No clinically significant adverse events occurred. In 26.8% of the bi-weekly surveys owners whose dogs received rapamycin reported perceived positive changes in behavior or health, compared to 8.1% in the placebo group (p = 0.04). Discussion While no clinically significant change in cardiac function was observed in dogs treated with low-dose rapamycin, the drug was well-tolerated with no significant adverse events.
Collapse
Affiliation(s)
- Brian G. Barnett
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Sonya R. Wesselowski
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Sonya G. Gordon
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Ashley B. Saunders
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Daniel E. L. Promislow
- Department of Biology, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Stephen M. Schwartz
- Epidemiology Program, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Lucy Chou
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Jeremy B. Evans
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Matt Kaeberlein
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Kate E. Creevy
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
6
|
Yan J, Feng G, Yang Y, Ding D, Ma L, Zhao X, Chen X, Wang H, Chen Z, Jin Q. Autophagy attenuates osteoarthritis in mice by inhibiting chondrocyte pyroptosis and improving subchondral bone remodeling. BIOMOLECULES AND BIOMEDICINE 2023; 23:77-88. [PMID: 35880352 PMCID: PMC9901906 DOI: 10.17305/bjbms.2022.7677] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/01/2022] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) is an age-related degenerative disease characterized by cartilage degeneration and abnormal bone remodeling in the subchondral bone. Autophagy maintains cellular homeostasis by self-phagocytosis. However, the underlying mechanisms of autophagy on the pathological progression of OA are still unknown. This study assessed the effects of autophagy on cartilage and subchondral bone in a mouse OA model. A mouse OA model was induced using destabilization of the medial meniscus (DMM) surgery. Assessment was performed by histomorphology, microcomputed tomography (micro-CT), immunohistochemical, immunofluorescent, and tartrate-resistant acid phosphatase (TRAP) staining. Our data revealed that autophagy can significantly delay the pathological progression of OA by increasing the thickness of hyaline cartilage and decreasing the thickness of calcified cartilage, increasing the subchondral bone volume fraction and bone mineralization density, and decreasing trabecular separation in the early stages of OA (2 weeks), whereas the opposite is true in the late stages of OA (8 weeks). Mechanistically, activation of autophagy in cartilage increased the expression of type II collagen (Col II), decreased the expression of matrix metalloproteinase 13 (MMP 13) and decreased the pyroptosis mediated by NOD-like receptor protein 3 (NLRP3) inflammasome by decreasing the expression of NLRP3, caspase-1, gasdermin D (GSDMD), and IL-1β. In the subchondral bone, activation of autophagy decreased the generation of mature osteoclasts at the early stages of OA (2 weeks) mainly by reducing the receptor activator for nuclear factor-κB ligand (RANKL)/osteoprotegerin (OPG) ratio, while it decreased osteoblastogenesis by reducing Runt-related transcription factor 2 (Runx2) expression significantly in the late stages of OA (8 weeks). In conclusion, autophagy may delay the pathological progression of OA in mice by inhibiting chondrocyte pyroptosis and improving subchondral bone remodeling.
Collapse
Affiliation(s)
- Jiangbo Yan
- Clinical College, Ningxia Medical University, Yinchuan, China,Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, China
| | - Gangning Feng
- Clinical College, Ningxia Medical University, Yinchuan, China
| | - Yong Yang
- Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, China
| | - Dong Ding
- Hand and Ankle Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Long Ma
- Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, China
| | - Xin Zhao
- Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, China
| | - Xiaolei Chen
- Clinical College, Ningxia Medical University, Yinchuan, China,Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, China
| | - Hui Wang
- Clinical College, Ningxia Medical University, Yinchuan, China
| | - Zhirong Chen
- Clinical College, Ningxia Medical University, Yinchuan, China,Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, China,Correspondence to Zhirong Chen: ; Qunhua Jin:
| | - Qunhua Jin
- Clinical College, Ningxia Medical University, Yinchuan, China,Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, China,Correspondence to Zhirong Chen: ; Qunhua Jin:
| |
Collapse
|
7
|
Wang J, Chen S, Pan C, Li G, Tang Z. Application of Small Molecules in the Central Nervous System Direct Neuronal Reprogramming. Front Bioeng Biotechnol 2022; 10:799152. [PMID: 35875485 PMCID: PMC9301571 DOI: 10.3389/fbioe.2022.799152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
The lack of regenerative capacity of neurons leads to poor prognoses for some neurological disorders. The use of small molecules to directly reprogram somatic cells into neurons provides a new therapeutic strategy for neurological diseases. In this review, the mechanisms of action of different small molecules, the approaches to screening small molecule cocktails, and the methods employed to detect their reprogramming efficiency are discussed, and the studies, focusing on neuronal reprogramming using small molecules in neurological disease models, are collected. Future research efforts are needed to investigate the in vivo mechanisms of small molecule-mediated neuronal reprogramming under pathophysiological states, optimize screening cocktails and dosing regimens, and identify safe and effective delivery routes to promote neural regeneration in different neurological diseases.
Collapse
Affiliation(s)
| | | | | | - Gaigai Li
- *Correspondence: Gaigai Li, ; Zhouping Tang,
| | | |
Collapse
|
8
|
Li Y, Zhao Y, Lu Y, Lu X, Hu Y, Li Q, Shuai M, Li R. Autism spectrum disorder-like behavior induced in rat offspring by perinatal exposure to di-(2-ethylhexyl) phthalate. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:52083-52097. [PMID: 35254616 DOI: 10.1007/s11356-022-19531-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 02/26/2022] [Indexed: 06/14/2023]
Abstract
Autism spectrum disorders (ASD), also known as childhood autism, is a common neurological developmental disorder. Although it is generally believed that genetic factors are a primary cause for ASD development, more and more studies show that an increasing number of ASD diagnoses are related to environmental exposure. Epidemiological studies indicated that perinatal exposure to endocrine disruptors might cause neurodevelopmental disorders in children. Di-(2-ethylhexyl) phthalate (DEHP) is widely used as a plasticizer in many products. To explore the neurodevelopmental effect induced by perinatal exposure to DEHP on rat offspring, and the potential mechanisms, female Wistar rats were exposed to 1, 10, and 100 mg/kg/day DEHP during pregnancy and lactation, while valproic acid (VPA) was used as a positive control. The behavior tests showed that rat pups exposed to VPA and 100 mg/kg/day DEHP were not good as those from the control group in both their socialability and social novelty. Expression of mTOR pathway-related components increased while the number of autophagosomes decreased in the brain tissue of the rat offspring exposed to 100 mg/kg/day DEHP. In addition, perinatal exposure to DEHP at all dosages decreased the level of autophagy proteins LC3II and Beclin1 in the brain tissue of rat pups. Our results indicated that perinatal DEHP exposure would induce ASD-like behavioral changes in rat offspring, which might be mediated by activation of the mTOR signaling pathway, and inhibition of autophagy in the brain.
Collapse
Affiliation(s)
- Yao Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
- Office of the Youth League Committee, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yun Zhao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Yu Lu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xianxian Lu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Yingdan Hu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Qiulin Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Menglei Shuai
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Rui Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, People's Republic of China.
| |
Collapse
|
9
|
Mohamed MA, Elkhateeb WA, Daba GM. Rapamycin golden jubilee and still the miraculous drug: a potent immunosuppressant, antitumor, rejuvenative agent, and potential contributor in COVID-19 treatment. BIORESOUR BIOPROCESS 2022; 9:65. [PMID: 35730039 PMCID: PMC9188914 DOI: 10.1186/s40643-022-00554-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/27/2022] [Indexed: 11/10/2022] Open
Abstract
Although celebrating its golden jubilee, rapamycin’s importance keeps increasing by the day. Starting as a promising antifungal agent, then as a potent immunosuppressant, strong anticancer drug, and now rapamycin is attracting serious attention as a rejuvenative agent and a possible contributor in treating this era pandemic, COVID-19. Due to its diverse biological activities and promising medical applications, we aimed in this review to put rapamycin under the spot and highlight its discovery, famous microbial producers, reported biological activities, chemical structure, famous analogues, and biosynthesis. Moreover, discuss some rapamycin production approaches including solid-state fermentation, and stressing out producing strain. On the other hand, describe its action mechanism and trials to use it in treatment of COVID-19. Additionally, we highlighted some of the side effects accompanying its use, and describe some approaches reported to minimize these undesired effects. Finally, we report the current status of rapamycin and its analogues in global market, and discuss future prospects of this potent drug.
Collapse
Affiliation(s)
- Mohamed A Mohamed
- Chemistry of Natural and Microbial Products Department, Pharmaceutical Industries Researches Institute, National Research Centre, El Buhouth St. Dokki, Giza, 12622 Egypt
| | - Waill A Elkhateeb
- Chemistry of Natural and Microbial Products Department, Pharmaceutical Industries Researches Institute, National Research Centre, El Buhouth St. Dokki, Giza, 12622 Egypt
| | - Ghoson M Daba
- Chemistry of Natural and Microbial Products Department, Pharmaceutical Industries Researches Institute, National Research Centre, El Buhouth St. Dokki, Giza, 12622 Egypt
| |
Collapse
|
10
|
Liu JK. Antiaging agents: safe interventions to slow aging and healthy life span extension. NATURAL PRODUCTS AND BIOPROSPECTING 2022; 12:18. [PMID: 35534591 PMCID: PMC9086005 DOI: 10.1007/s13659-022-00339-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 03/29/2022] [Indexed: 05/02/2023]
Abstract
Human longevity has increased dramatically during the past century. More than 20% of the 9 billion population of the world will exceed the age of 60 in 2050. Since the last three decades, some interventions and many preclinical studies have been found to show slowing aging and increasing the healthy lifespan of organisms from yeast, flies, rodents to nonhuman primates. The interventions are classified into two groups: lifestyle modifications and pharmacological/genetic manipulations. Some genetic pathways have been characterized to have a specific role in controlling aging and lifespan. Thus, all genes in the pathways are potential antiaging targets. Currently, many antiaging compounds target the calorie-restriction mimetic, autophagy induction, and putative enhancement of cell regeneration, epigenetic modulation of gene activity such as inhibition of histone deacetylases and DNA methyltransferases, are under development. It appears evident that the exploration of new targets for these antiaging agents based on biogerontological research provides an incredible opportunity for the healthcare and pharmaceutical industries. The present review focus on the properties of slow aging and healthy life span extension of natural products from various biological resources, endogenous substances, drugs, and synthetic compounds, as well as the mechanisms of targets for antiaging evaluation. These bioactive compounds that could benefit healthy aging and the potential role of life span extension are discussed.
Collapse
Affiliation(s)
- Ji-Kai Liu
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, People's Republic of China.
| |
Collapse
|
11
|
Gibbs NH, Michalski H, Promislow DEL, Kaeberlein M, Creevy KE. Reasons for Exclusion of Apparently Healthy Mature Adult and Senior Dogs From a Clinical Trial. Front Vet Sci 2021; 8:651698. [PMID: 34150883 PMCID: PMC8206478 DOI: 10.3389/fvets.2021.651698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 04/27/2021] [Indexed: 11/14/2022] Open
Abstract
Background: Interventional clinical trials intended to maintain health in aging dogs are unusual and require particular attention to exclusion criteria. Objectives: To describe reasons for exclusion when a mature adult and senior canine population with normal health status was sought. Animals: Fifty six companion dogs nominated for a randomized controlled trial (RCT). Procedures: Exclusions occurred within Stage 1 (S1): owner-provided survey information; Stage 2 (S2): medical records review; and Stage 3 (S3): screening examination and within Owner, Dog, or Other factor categories. Results: Of 56 nominated dogs, 39 were excluded at S1 (n = 19), S2 (n = 5), and S3 (n = 15), respectively. Dogs were excluded for Owner (n = 4), Dog (n = 27), Other (n = 6), and concurrent (Owner + Dog; n = 2) factors. The most common exclusion period was S1 (n = 19), with weight outside the target range being the most common exclusion factor in that stage (n = 10). Heart murmurs were the second most common exclusion factor (S1: n = 1; S3: n = 5); suspected or confirmed systemic illness was third most common (S1: n = 2; S2: n = 3; S3: n = 2). Among dogs who passed S1 and S2 screening (n = 32), 15 dogs (48%) were excluded at S3, for heart murmur > grade II/VI (n = 5), cardiac arrhythmias (n = 2), and clinicopathologic abnormalities (n = 2). Conclusions and Clinical Relevance: Dogs nominated for a clinical trial for healthy mature adult and senior dogs were excluded for size, previous diagnoses, and newly discovered cardiac abnormalities. For future interventions in mature adult and senior dogs of normal health status, it is important to define expected age-related abnormalities to ensure that meaningful exclusion criteria are used.
Collapse
Affiliation(s)
- Nicole H Gibbs
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Hannah Michalski
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Daniel E L Promislow
- Department of Laboratory Medicine & Pathology, University of Washington School of Medicine, Seattle, WA, United States.,Department of Biology, University of Washington, Seattle, WA, United States
| | - Matt Kaeberlein
- Department of Laboratory Medicine & Pathology, University of Washington School of Medicine, Seattle, WA, United States
| | - Kate E Creevy
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
12
|
Alterations in Tau Protein Level and Phosphorylation State in the Brain of the Autistic-Like Rats Induced by Prenatal Exposure to Valproic Acid. Int J Mol Sci 2021; 22:ijms22063209. [PMID: 33809910 PMCID: PMC8004207 DOI: 10.3390/ijms22063209] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/23/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficient social interaction and communication besides repetitive, stereotyped behaviours. A characteristic feature of ASD is altered dendritic spine density and morphology associated with synaptic plasticity disturbances. Since microtubules (MTs) regulate dendritic spine morphology and play an important role in spine development and plasticity the aim of the present study was to investigate the alterations in the content of neuronal α/β-tubulin and Tau protein level as well as phosphorylation state in the valproic acid (VPA)-induced rat model of autism. Our results indicated that maternal exposure to VPA induces: (1) decrease the level of α/β-tubulin along with Tau accumulation in the hippocampus and cerebral cortex; (2) excessive Tau phosphorylation and activation of Tau-kinases: CDK5, ERK1/2, and p70S6K in the cerebral cortex; (3) up-regulation of mTOR kinase-dependent signalling in the hippocampus and cerebral cortex of adolescent rat offspring. Moreover, immunohistochemical staining showed histopathological changes in neurons (chromatolysis) in both analysed brain structures of rats prenatally exposed to VPA. The observed changes in Tau protein together with an excessive decrease in α/β-tubulin level may suggest destabilization and thus dysfunction of the MT cytoskeleton network, which in consequence may lead to the disturbance in synaptic plasticity and the development of autistic-like behaviours.
Collapse
|
13
|
The potential of rapalogs to enhance resilience against SARS-CoV-2 infection and reduce the severity of COVID-19. LANCET HEALTHY LONGEVITY 2021; 2:e105-e111. [PMID: 33665645 PMCID: PMC7906698 DOI: 10.1016/s2666-7568(20)30068-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
COVID-19 disproportionately affects older people, with likelihood of severe complications and death mirroring that of other age-associated diseases. Inhibition of the mechanistic target of rapamycin complex 1 (mTORC1) has been shown to delay or reverse many age-related phenotypes, including declining immune function. Rapamycin (sirolimus) and rapamycin derivatives are US Food and Drug Administration-approved inhibitors of mTORC1 with broad clinical utility and well established dosing and safety profiles. Based on preclinical and clinical evidence, a strong case can be made for immediate large-scale clinical trials to assess whether rapamycin and other mTORC1 inhibitors can prevent COVID-19 infection in these populations and also to determine whether these drugs can improve outcomes in patients with severe COVID-19.
Collapse
|
14
|
Duan Q, Li S, Wen X, Sunnassee G, Chen J, Tan S, Guo Y. Valproic Acid Enhances Reprogramming Efficiency and Neuronal Differentiation on Small Molecules Staged-Induction Neural Stem Cells: Suggested Role of mTOR Signaling. Front Neurosci 2019; 13:867. [PMID: 31551670 PMCID: PMC6737087 DOI: 10.3389/fnins.2019.00867] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/02/2019] [Indexed: 12/21/2022] Open
Abstract
Inducing somatic cells into neural stem cells (iNSCs) in specific ways provides a new cell therapy in a variety of neurological diseases. In the past, iNSCs were generated by transcription factors which increased the risk of mutagenesis, tumor formations, and immune reactions by viral transduction vectors. Therefore, in this study, different small molecules were used to induce mouse embryonic fibroblasts (MEFs) into iNSCs in different reprogramming stages, which showed high reprogramming efficiency without altering the genome. We demonstrated that the small molecules staged-induction neural stem cells (SMSINS) have the characteristics of neural stem cells (NSCs) in morphology, gene expression, self-renewal and differentiation potential. Furthermore, valproic acid (VPA), one of small molecules, was showed to enhance neural induction with highest efficiency compared with six other small molecules, which were also investigated in the present study. Moreover, our results suggested that activating the mammalian target of rapamycin (mTOR) signaling enhanced the induction efficiency and neuronal differentiation. Collectively, our findings indicated that using this induction program allowed us to obtain safe and efficient iNSCs which were free of genetic manipulation. The VPA-mediated mTOR signaling pathway may enhance reprogramming efficiency and neuronal differentiation. So we suggested that this program could be a new method of obtaining iNSCs for the treatment of neurological diseases by cell replacement therapy in the future.
Collapse
Affiliation(s)
- Qingrui Duan
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Siyi Li
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xinrui Wen
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Gavin Sunnassee
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Chen
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Sheng Tan
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Guo
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Escobar KA, Cole NH, Mermier CM, VanDusseldorp TA. Autophagy and aging: Maintaining the proteome through exercise and caloric restriction. Aging Cell 2019; 18:e12876. [PMID: 30430746 PMCID: PMC6351830 DOI: 10.1111/acel.12876] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 08/31/2018] [Accepted: 09/28/2018] [Indexed: 12/16/2022] Open
Abstract
Accumulation of dysfunctional and damaged cellular proteins and organelles occurs during aging, resulting in a disruption of cellular homeostasis and progressive degeneration and increases the risk of cell death. Moderating the accrual of these defunct components is likely a key in the promotion of longevity. While exercise is known to promote healthy aging and mitigate age‐related pathologies, the molecular underpinnings of this phenomenon remain largely unclear. However, recent evidences suggest that exercise modulates the proteome. Similarly, caloric restriction (CR), a known promoter of lifespan, is understood to augment intracellular protein quality. Autophagy is an evolutionary conserved recycling pathway responsible for the degradation, then turnover of cellular proteins and organelles. This housekeeping system has been reliably linked to the aging process. Moreover, autophagic activity declines during aging. The target of rapamycin complex 1 (TORC1), a central kinase involved in protein translation, is a negative regulator of autophagy, and inhibition of TORC1 enhances lifespan. Inhibition of TORC1 may reduce the production of cellular proteins which may otherwise contribute to the deleterious accumulation observed in aging. TORC1 may also exert its effects in an autophagy‐dependent manner. Exercise and CR result in a concomitant downregulation of TORC1 activity and upregulation of autophagy in a number of tissues. Moreover, exercise‐induced TORC1 and autophagy signaling share common pathways with that of CR. Therefore, the longevity effects of exercise and CR may stem from the maintenance of the proteome by balancing the synthesis and recycling of intracellular proteins and thus may represent practical means to promote longevity.
Collapse
Affiliation(s)
- Kurt A. Escobar
- Department of Kinesiology; California State University, Long Beach; Long Beach California
| | - Nathan H. Cole
- Department of Health, Exercise, & Sports Sciences; University of New Mexico; Albuquerque New Mexico
| | - Christine M. Mermier
- Department of Health, Exercise, & Sports Sciences; University of New Mexico; Albuquerque New Mexico
| | - Trisha A. VanDusseldorp
- Department of Exercise Science & Sports Management; Kennesaw State University; Kennesaw Georgia
| |
Collapse
|
16
|
Wang L, Cai Y, Fan X. Metformin Administration During Early Postnatal Life Rescues Autistic-Like Behaviors in the BTBR T+ Itpr3tf/J Mouse Model of Autism. Front Behav Neurosci 2018; 12:290. [PMID: 30555309 PMCID: PMC6281763 DOI: 10.3389/fnbeh.2018.00290] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 11/09/2018] [Indexed: 01/21/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disability characterized by impaired social interactions, stereotypical repetitive behavior and restricted interests. Although the global incidence of ASD has increased over time, the etiology of ASD is poorly understood, and there is no effective pharmacological intervention for treating ASD. Recent studies have suggested that metformin has the potential to treat ASD. Thus, in this study, we assessed the therapeutic effects of early metformin treatment in a BTBR T+ Itpr3tf/J (BTBR) mouse model of ASD. We observed that early metformin administration significantly reversed social approach deficits, attenuated repetitive grooming and reduced marble burying in BTBR mice. Metformin did not change the general locomotor activity or anxiety-like behavior in both BTBR and C57BL/6J (B6) mice. Our findings suggest that early metformin treatment may have beneficial effects on ameliorating behavioral deficits in ASD.
Collapse
Affiliation(s)
- Lian Wang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Yulong Cai
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, China
| |
Collapse
|
17
|
Wang A, Mouser J, Pitt J, Promislow D, Kaeberlein M. Rapamycin enhances survival in a Drosophila model of mitochondrial disease. Oncotarget 2018; 7:80131-80139. [PMID: 27741510 PMCID: PMC5348310 DOI: 10.18632/oncotarget.12560] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/27/2016] [Indexed: 11/25/2022] Open
Abstract
Pediatric mitochondrial disorders are a devastating category of diseases caused by deficiencies in mitochondrial function. Leigh Syndrome (LS) is the most common of these diseases with symptoms typically appearing within the first year of birth and progressing rapidly until death, usually by 6-7 years of age. Our lab has recently shown that genetic inhibition of the mechanistic target of rapamycin (TOR) rescues the short lifespan of yeast mutants with defective mitochondrial function, and that pharmacological inhibition of TOR by administration of rapamycin significantly rescues the shortened lifespan, neurological symptoms, and neurodegeneration in a mouse model of LS. However, the mechanism by which TOR inhibition exerts these effects, and the extent to which these effects can extend to other models of mitochondrial deficiency, are unknown. Here, we probe the effects of TOR inhibition in a Drosophila model of complex I deficiency. Treatment with rapamycin robustly suppresses the lifespan defect in this model of LS, without affecting behavioral phenotypes. Interestingly, this increased lifespan in response to TOR inhibition occurs in an autophagy-independent manner. Further, we identify a fat storage defect in the ND2 mutant flies that is rescued by rapamycin, supporting a model that rapamycin exerts its effects on mitochondrial disease in these animals by altering metabolism.
Collapse
Affiliation(s)
- Adrienne Wang
- University of Washington, Department of Pathology, Seattle, WA, USA
| | - Jacob Mouser
- University of Washington, Department of Pathology, Seattle, WA, USA
| | - Jason Pitt
- University of Washington, Department of Pathology, Seattle, WA, USA
| | - Daniel Promislow
- University of Washington, Department of Pathology, Seattle, WA, USA.,University of Washington, Department of Biology, Seattle, WA, USA
| | - Matt Kaeberlein
- University of Washington, Department of Pathology, Seattle, WA, USA
| |
Collapse
|
18
|
Rapamycin in aging and disease: maximizing efficacy while minimizing side effects. Oncotarget 2018; 7:44876-44878. [PMID: 27384492 PMCID: PMC5216691 DOI: 10.18632/oncotarget.10381] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/16/2016] [Indexed: 12/31/2022] Open
Abstract
Experimental geroscience has identified rapamycin as a top candidate for promoting healthy aging and longevity in mammals. As multiple independent studies have successfully reproduced the lifespan and healthspan promoting effects of rapamycin, the focus has shifted to possible translational use. While a promising compound, clinical use of rapamycin is limited by concerns of side effects associated with the drug. Studies aimed at defining optimal dosage regimen, delivery route, and formulation will allow for benefits to be maximized while reducing side effects.
Collapse
|
19
|
Wang L, Cai Y, Fan X. Metformin Administration During Early Postnatal Life Rescues Autistic-Like Behaviors in the BTBR T+ Itpr3tf/J Mouse Model of Autism. Front Behav Neurosci 2018; 12:290. [PMID: 30555309 DOI: 10.3389/fnbeh.2018.00290/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 11/09/2018] [Indexed: 05/20/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disability characterized by impaired social interactions, stereotypical repetitive behavior and restricted interests. Although the global incidence of ASD has increased over time, the etiology of ASD is poorly understood, and there is no effective pharmacological intervention for treating ASD. Recent studies have suggested that metformin has the potential to treat ASD. Thus, in this study, we assessed the therapeutic effects of early metformin treatment in a BTBR T+ Itpr3tf/J (BTBR) mouse model of ASD. We observed that early metformin administration significantly reversed social approach deficits, attenuated repetitive grooming and reduced marble burying in BTBR mice. Metformin did not change the general locomotor activity or anxiety-like behavior in both BTBR and C57BL/6J (B6) mice. Our findings suggest that early metformin treatment may have beneficial effects on ameliorating behavioral deficits in ASD.
Collapse
Affiliation(s)
- Lian Wang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Yulong Cai
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, China
| |
Collapse
|
20
|
Kaeberlein M. Translational geroscience: A new paradigm for 21 st century medicine. TRANSLATIONAL MEDICINE OF AGING 2017; 1:1-4. [PMID: 32219192 DOI: 10.1016/j.tma.2017.09.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Advances in geroscience are allowing scientists and clinicians, for the first time, to consider interventions aimed at directly targeting the hallmarks of aging. Unlike disease-specific approaches, such interventions have the potential to prevent multiple diseases of aging simultaneously, thereby greatly enhancing healthspan for most individuals. Initial clinical data indicates that geroprotective compounds such as rapamycin and metformin may be effective at delaying or reversing age-related disease in otherwise healthy elderly people and companion animals. Here I will provide an overview of the field of translational geroscience, which I believe will become the paradigm for the practice of medicine in the 21st century.
Collapse
Affiliation(s)
- Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, WA, 98195-7470, USA
| |
Collapse
|
21
|
An JY, Quarles EK, Mekvanich S, Kang A, Liu A, Santos D, Miller RA, Rabinovitch PS, Cox TC, Kaeberlein M. Rapamycin treatment attenuates age-associated periodontitis in mice. GeroScience 2017; 39:457-463. [PMID: 28889220 PMCID: PMC5636779 DOI: 10.1007/s11357-017-9994-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 08/17/2017] [Indexed: 01/07/2023] Open
Abstract
Interventions that target biological mechanisms of aging have great potential to enhance quality of life by delaying morbidity and mortality. The FDA-approved drug rapamycin is a compelling candidate for such an intervention. In a previous study, it was reported that 3 months of rapamycin treatment is sufficient to increase life expectancy and remodel the gut microbiome in aged mice. Transient treatment with rapamycin or a rapamycin derivative has also been shown to delay immune stem cell senescence and rejuvenate immune function in aged mice and elderly people. Periodontal disease is an important age-related disease involving altered immune function, pathological changes to the oral microbiome, and systemic inflammation. Periodontal disease is defined clinically by loss of alveolar bone and by connective tissue degeneration. Here, we describe significant alveolar bone loss during aging in two different mouse strain backgrounds and report that rapamycin treatment is sufficient to reverse age-associated periodontal disease in mice. Partial restoration of youthful levels of alveolar bone is observed in 22-month-old rapamycin-treated mice as rapidly as 8 weeks after initiation of treatment. To the best of our knowledge, this represents the first intervention shown to substantially prevent or reverse age-associated alveolar bone loss. These findings suggest the possibility that inhibition of mTOR with rapamycin or other pharmacological agents may be useful to treat a clinically relevant condition for which there is currently no effective treatment.
Collapse
Affiliation(s)
- Jonathan Y An
- Department of Oral Health Sciences, University of Washington School of Dentistry, Seattle, WA, 98195, USA
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Ellen K Quarles
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Surapat Mekvanich
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Alex Kang
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Anthony Liu
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Danielle Santos
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Peter S Rabinovitch
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Timothy C Cox
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA
| | - Matt Kaeberlein
- Department of Oral Health Sciences, University of Washington School of Dentistry, Seattle, WA, 98195, USA.
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
22
|
A system to identify inhibitors of mTOR signaling using high-resolution growth analysis in Saccharomyces cerevisiae. GeroScience 2017; 39:419-428. [PMID: 28707282 DOI: 10.1007/s11357-017-9988-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 06/27/2017] [Indexed: 10/19/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) is a central regulator of growth and proliferation and mTOR inhibition is a promising therapy for a variety of diseases and disorders. Inhibition of mTOR complex I (mTORC1) with rapamycin delays aging and increases healthy longevity in laboratory animals and is used clinically at high doses to prevent organ transplant rejection and to treat some forms of cancer. Clinical use of rapamycin is associated with several unwanted side effects, however, and several strategies are being taken to identify mTORC1 inhibitors with fewer side effects. We describe here a yeast-based growth assay that can be used to screen for novel inhibitors of mTORC1. By testing compounds using a wild-type strain and isogenic cells lacking either TOR1 or FPR1, we can resolve not only whether a compound is an inhibitor of mTORC1 but also whether the inhibitor acts through a mechanism similar to rapamycin by binding Fpr1. Using this assay, we show that rapamycin derivatives behave similarly to rapamycin, while caffeine and the ATP competitive inhibitors Torin 1 and GSK2126458 are mTORC1 inhibitors in yeast that act independently of Fpr1. Some mTOR inhibitors in mammalian cells do not inhibit mTORC1 in yeast, and several nutraceutical compounds were not found to specifically inhibit mTOR but resulted in a general inhibition of yeast growth. Our screening method holds promise as a means of effectively assaying drug libraries for mTOR-inhibitory molecules in vivo that may be adapted as novel treatments to fight diseases and extend healthy longevity.
Collapse
|
23
|
A randomized controlled trial to establish effects of short-term rapamycin treatment in 24 middle-aged companion dogs. GeroScience 2017; 39:117-127. [PMID: 28374166 DOI: 10.1007/s11357-017-9972-z] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 03/24/2017] [Indexed: 01/19/2023] Open
Abstract
Age is the single greatest risk factor for most causes of morbidity and mortality in humans and their companion animals. As opposed to other model organisms used to study aging, dogs share the human environment, are subject to similar risk factors, receive comparable medical care, and develop many of the same age-related diseases humans do. In this study, 24 middle-aged healthy dogs received either placebo or a non-immunosuppressive dose of rapamycin for 10 weeks. All dogs received clinical and hematological exams before, during, and after the trial and echocardiography before and after the trial. Our results showed no clinical side effects in the rapamycin-treated group compared to dogs receiving the placebo. Echocardiography suggested improvement in both diastolic and systolic age-related measures of heart function (E/A ratio, fractional shortening, and ejection fraction) in the rapamycin-treated dogs. Hematological values remained within the normal range for all parameters studied; however, the mean corpuscular volume (MCV) was decreased in rapamycin-treated dogs. Based on these results, we will test rapamycin on a larger dog cohort for a longer period of time in order to validate its effects on cardiac function and to determine whether it can significantly improve healthspan and reduce mortality in companion dogs.
Collapse
|
24
|
Haubrich BA, Swinney DC. Enzyme Activity Assays for Protein Kinases: Strategies to Identify Active Substrates. Curr Drug Discov Technol 2016; 13:2-15. [PMID: 26768716 DOI: 10.2174/1570163813666160115125930] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/13/2016] [Accepted: 01/13/2016] [Indexed: 11/22/2022]
Abstract
Protein kinases are an important class of enzymes and drug targets. New opportunities to discover medicines for neglected diseases can be leveraged by the extensive kinase tools and knowledge created in targeting human kinases. A valuable tool for kinase drug discovery is an enzyme assay that measures catalytic function. The functional assay can be used to identify inhibitors, estimate affinity, characterize molecular mechanisms of action (MMOAs) and evaluate selectivity. However, establishing an enzyme assay for a new kinases requires identification of a suitable substrate. Identification of a new kinase's endogenous physiologic substrate and function can be extremely costly and time consuming. Fortunately, most kinases are promiscuous and will catalyze the phosphotransfer from ATP to alternative substrates with differing degrees of catalytic efficiency. In this manuscript we review strategies and successes in the identification of alternative substrates for kinases from organisms responsible for many of the neglected tropical diseases (NTDs) towards the goal of informing strategies to identify substrates for new kinases. Approaches for establishing a functional kinase assay include measuring auto-activation and use of generic substrates and peptides. The most commonly used generic substrates are casein, myelin basic protein, and histone. Sequence homology modeling can provide insights into the potential substrates and the requirement for activation. Empirical approaches that can identify substrates include screening of lysates (which may also help identify native substrates) and use of peptide arrays. All of these approaches have been used with a varying degree of success to identify alternative substrates.
Collapse
Affiliation(s)
- Brad A Haubrich
- Institute for Rare and Neglected Diseases Drug Discovery, 897 Independence Ave, Suite 2C, Mountain View, CA 94043, USA.
| | | |
Collapse
|
25
|
Pazoki-Toroudi H, Amani H, Ajami M, Nabavi SF, Braidy N, Kasi PD, Nabavi SM. Targeting mTOR signaling by polyphenols: A new therapeutic target for ageing. Ageing Res Rev 2016; 31:55-66. [PMID: 27453478 DOI: 10.1016/j.arr.2016.07.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 06/19/2016] [Accepted: 07/15/2016] [Indexed: 12/19/2022]
Abstract
Current ageing research is aimed not only at the promotion of longevity, but also at improving health span through the discovery and development of new therapeutic strategies by investigating molecular and cellular pathways involved in cellular senescence. Understanding the mechanism of action of polyphenolic compounds targeting mTOR (mechanistic target of rapamycin) and related pathways opens up new directions to revolutionize ways to slow down the onset and development of age-dependent degeneration. Herein, we will discuss the mechanisms by which polyphenols can delay the molecular pathogenesis of ageing via manipulation or more specifically inhibition of mTOR-signaling pathways. We will also discuss the implications of polyphenols in targeting mTOR and its related pathways on health life span extension and longevity..
Collapse
|
26
|
Tramutola A, Lanzillotta C, Di Domenico F. Targeting mTOR to reduce Alzheimer-related cognitive decline: from current hits to future therapies. Expert Rev Neurother 2016; 17:33-45. [PMID: 27690737 DOI: 10.1080/14737175.2017.1244482] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The mTOR pathway is involved in the regulation of a wide repertoire of cellular functions in the brain and its dysregulation is emerging as a leitmotif in a large number of neurological disorders. In AD, altered mTOR signaling contributes to the inhibition of autophagy deposition of Aβ and tau aggregates and to the alteration of several neuronal metabolic pathways. Areas covered: In this review, we report all the current findings on the use of mTOR inhibitors (rapamycin, rapalogues) in the treatment of AD. These results support the role of mTOR inhibitors as potential therapeutic agents able to reduce AD hallmarks and recover cognitive performances. Expert commentary: Despite mTOR inhibitors appearing to be ideal compounds to counteract AD, further studies are needed in order to gain knowledge on the involvement of aberrant mTOR in AD, and to standardize a valuable therapeutic approach that can be translated to humans.
Collapse
Affiliation(s)
- Antonella Tramutola
- a Department of Biochemical Sciences , Sapienza University of Rome , Rome , Italy
| | - Chiara Lanzillotta
- a Department of Biochemical Sciences , Sapienza University of Rome , Rome , Italy
| | - Fabio Di Domenico
- a Department of Biochemical Sciences , Sapienza University of Rome , Rome , Italy
| |
Collapse
|
27
|
Qin L, Dai X, Yin Y. Valproic acid exposure sequentially activates Wnt and mTOR pathways in rats. Mol Cell Neurosci 2016; 75:27-35. [PMID: 27343825 DOI: 10.1016/j.mcn.2016.06.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 05/11/2016] [Accepted: 06/21/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impaired social interaction, limited verbal communication and repetitive behaviors. Recent studies have demonstrated that Wnt signaling and mTOR signaling play important roles in the pathogenesis of ASD. However, the relationship of these two signaling pathways in ASD remains unclear. RESULTS We assessed this question using the valproic acid (VPA) rat model of autism. Our results demonstrated that VPA exposure activated mTOR signaling and suppressed autophagy in the prefrontal cortex, hippocampus and cerebellum of autistic model rats, characterized by enhanced phospho-mTOR and phospho-S6 and decreased Beclin1, Atg5, Atg10, LC3-II and autophagosome formation. Rapamycin treatment suppressed the effect of VPA on mTOR signaling and ameliorated the autistic-like behaviors of rats in our autism model. The administration of VPA also activated Wnt signaling through up-regulating beta-catenin and phospho-GSK3beta. Suppression of the Wnt pathway by sulindac relieved autistic-like behaviors and attenuated VPA-induced mTOR signaling activation in autistic model rats. CONCLUSIONS Our results demonstrate that VPA exposure sequentially activates Wnt signaling and mTOR signaling in rats. Suppression of the Wnt signaling pathway relieves autistic-like behaviors partially by deactivating the mTOR signaling pathway in VPA-exposed rats.
Collapse
Affiliation(s)
- Liyan Qin
- Chongqing Key Laboratory of Psychological Diagnosis and Educational Technology for Children with Special Needs, Chongqing 400047, China; Department of Blood Transfusion, Southwest Hospital of Third Military Medical University,Chongqing 400038, China
| | - Xufang Dai
- Chongqing Key Laboratory of Psychological Diagnosis and Educational Technology for Children with Special Needs, Chongqing 400047, China; College of Education Science, Chongqing Normal University, Chongqing 400047, China.
| | - Yunhou Yin
- Guizhou Minzu University, Guiyang 550025, China
| |
Collapse
|
28
|
Kaeberlein M, Creevy KE, Promislow DEL. The dog aging project: translational geroscience in companion animals. Mamm Genome 2016; 27:279-88. [PMID: 27143112 DOI: 10.1007/s00335-016-9638-7] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/15/2016] [Indexed: 12/16/2022]
Abstract
Studies of the basic biology of aging have identified several genetic and pharmacological interventions that appear to modulate the rate of aging in laboratory model organisms, but a barrier to further progress has been the challenge of moving beyond these laboratory discoveries to impact health and quality of life for people. The domestic dog, Canis familiaris, offers a unique opportunity for surmounting this barrier in the near future. In particular, companion dogs share our environment and play an important role in improving the quality of life for millions of people. Here, we present a rationale for increasing the role of companion dogs as an animal model for both basic and clinical geroscience and describe complementary approaches and ongoing projects aimed at achieving this goal.
Collapse
Affiliation(s)
- Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, WA, USA.
| | - Kate E Creevy
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | | |
Collapse
|
29
|
Chronic cerebral hypoperfusion enhances Tau hyperphosphorylation and reduces autophagy in Alzheimer's disease mice. Sci Rep 2016; 6:23964. [PMID: 27050297 PMCID: PMC4822118 DOI: 10.1038/srep23964] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/14/2016] [Indexed: 12/13/2022] Open
Abstract
Cerebral hypoperfusion and impaired autophagy are two etiological factors that have been identified as being associated with the development of Alzheimer’s disease (AD). Nevertheless, the exact relationships among these pathological processes remain unknown. To elucidate the impact of cerebral hypoperfusion in AD, we created a unilateral common carotid artery occlusion (UCCAO) model by occluding the left common carotid artery in both young and old 3xTg-AD mice. Two months after occlusion, we found that ligation increases phospho-Tau (p-Tau) at Serine 199/202 in the hippocampus of 3-month-old AD mice, compared to sham-operated AD mice; whereas, there is no change in the wild type (WT) mice after ligation. Moreover, cerebral hypoperfusion led to significant increase of p-Tau in both the hippocampus and cortex of 16-month-old AD mice and WT mice. Notably, we did not detect any change in Aβ42 level in either young or old AD and WT mice after ligation. Interestingly, we observed a downregulation of LC3-II in the cortex of aged AD mice and WT mice after ligation. Our results suggest that elevated p-Tau and reduced autophagy are major cellular changes that are associated with hypoperfusion in AD. Therefore, targeting p-Tau and autophagy pathways may ameliorate hypoperfusion-induced brain damage in AD.
Collapse
|
30
|
Kaeberlein M. The Biology of Aging: Citizen Scientists and Their Pets as a Bridge Between Research on Model Organisms and Human Subjects. Vet Pathol 2016; 53:291-8. [PMID: 26077786 PMCID: PMC4794982 DOI: 10.1177/0300985815591082] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A fundamental goal of research into the basic mechanisms of aging is to develop translational strategies that improve human health by delaying the onset and progression of age-related pathology. Several interventions have been discovered that increase life span in invertebrate organisms, some of which have similar effects in mice. These include dietary restriction and inhibition of the mechanistic target of rapamycin by treatment with rapamycin. Key challenges moving forward will be to assess the extent to which these and other interventions improve healthy longevity and increase life span in mice and to develop practical strategies for extending this work to the clinic. Companion animals may provide an optimal intermediate between laboratory models and humans. By improving healthy longevity in companion animals, important insights will be gained regarding human aging while improving the quality of life for people and their pets.
Collapse
Affiliation(s)
- M Kaeberlein
- Department of Pathology, University of Washington, Seattle, WA, USA
| |
Collapse
|
31
|
Leontieva OV, Blagosklonny MV. Tumor promoter-induced cellular senescence: cell cycle arrest followed by geroconversion. Oncotarget 2015; 5:12715-27. [PMID: 25587030 PMCID: PMC4350340 DOI: 10.18632/oncotarget.3011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 12/26/2014] [Indexed: 02/07/2023] Open
Abstract
Phorbol ester (PMA or TPA), a tumor promoter, can cause either proliferation or cell cycle arrest, depending on cellular context. For example, in SKBr3 breast cancer cells, PMA hyper-activates the MEK/MAPK pathway, thus inducing p21 and cell cycle arrest. Here we showed that PMA-induced arrest was followed by conversion to cellular senescence (geroconversion). Geroconversion was associated with active mTOR and S6 kinase (S6K). Rapamycin suppressed geroconversion, maintaining quiescence instead. In this model, PMA induced arrest (step one of a senescence program), whereas constitutively active mTOR drove geroconversion (step two). Without affecting Akt phosphorylation, PMA increased phosphorylation of S6K (T389) and S6 (S240/244), and that was completely prevented by rapamycin. Yet, T421/S424 and S235/236 (p-S6K and p-S6, respectively) phosphorylation became rapamycin-insensitive in the presence of PMA. Either MEK or mTOR was sufficient to phosphorylate these PMA-induced rapamycin-resistant sites because co-treatment with U0126 and rapamycin was required to abrogate them. We next tested whether activation of rapamycin-insensitive pathways would shift quiescence towards senescence. In HT-p21 cells, cell cycle arrest was caused by IPTG-inducible p21 and was spontaneously followed by mTOR-dependent geroconversion. Rapamycin suppressed geroconversion, whereas PMA partially counteracted the effect of rapamycin, revealing the involvement of rapamycin-insensitive gerogenic pathways. In normal RPE cells arrested by serum withdrawal, the mTOR/pS6 pathway was inhibited and cells remained quiescent. PMA transiently activated mTOR, enabling partial geroconversion. We conclude that PMA can initiate a senescent program by either inducing arrest or fostering geroconversion or both. Rapamycin can decrease gero-conversion by PMA, without preventing PMA-induced arrest. The tumor promoter PMA is a gero-promoter, which may be useful to study aging in mammals.
Collapse
Affiliation(s)
- Olga V Leontieva
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | |
Collapse
|
32
|
Abstract
The most physiological type of cell cycle arrest - namely, contact inhibition in dense culture - is the least densely studied. Despite cell cycle arrest, confluent cells do not become senescent. We recently described that mTOR (target of rapamycin) is inactive in contact-inhibited cells. Therefore, conversion from reversible arrest to senescence (geroconversion) is suppressed. I this Perspective, we further extended the gerosuppression model. While causing senescence in regular cell density, etoposide failed to cause senescence in contact-inhibited cells. A transient reactivation of mTOR favored geroconversion in etoposide-treated confluent cells. Like p21, p16 did not cause senescence in high cell density. We discuss that suppression of geroconversion in confluent and contact-inhibited cultures mimics gerosuppression in the organism. We confirmed that levels of p-S6 were low in murine tissues in the organism compared with mouse embryonic fibroblasts in cell culture, whereas p-Akt was reciprocally high in the organism.
Collapse
Affiliation(s)
- Olga V Leontieva
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elms and Carlson Streets, Buffalo, NY 14263, USA
| | - Mikhail V Blagosklonny
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elms and Carlson Streets, Buffalo, NY 14263, USA
| |
Collapse
|
33
|
Johnson SC, Yanos ME, Bitto A, Castanza A, Gagnidze A, Gonzalez B, Gupta K, Hui J, Jarvie C, Johnson BM, Letexier N, McCanta L, Sangesland M, Tamis O, Uhde L, Van Den Ende A, Rabinovitch PS, Suh Y, Kaeberlein M. Dose-dependent effects of mTOR inhibition on weight and mitochondrial disease in mice. Front Genet 2015; 6:247. [PMID: 26257774 PMCID: PMC4510413 DOI: 10.3389/fgene.2015.00247] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/06/2015] [Indexed: 11/13/2022] Open
Abstract
Rapamycin extends lifespan and attenuates age-related pathologies in mice when administered through diet at 14 parts per million (PPM). Recently, we reported that daily intraperitoneal injection of rapamycin at 8 mg/kg attenuates mitochondrial disease symptoms and progression in the Ndufs4 knockout mouse model of Leigh Syndrome. Although rapamycin is a widely used pharmaceutical agent dosage has not been rigorously examined and no dose-response profile has been established. Given these observations we sought to determine if increased doses of oral rapamycin would result in more robust impact on mTOR driven parameters. To test this hypothesis, we compared the effects of dietary rapamycin at doses ranging from 14 to 378 PPM on developmental weight in control and Ndufs4 knockout mice and on health and survival in the Ndufs4 knockout model. High dose rapamycin was well tolerated, dramatically reduced weight gain during development, and overcame gender differences. The highest oral dose, approximately 27-times the dose shown to extend murine lifespan, increased survival in Ndufs4 knockout mice similarly to daily rapamycin injection without observable adverse effects. These findings have broad implications for the effective use of rapamycin in murine studies and for the translational potential of rapamycin in the treatment of mitochondrial disease. This data, further supported by a comparison of available literature, suggests that 14 PPM dietary rapamycin is a sub-optimal dose for targeting mTOR systemically in mice. Our findings suggest that the role of mTOR in mammalian biology may be broadly underestimated when determined through treatment with rapamycin at commonly used doses.
Collapse
Affiliation(s)
- Simon C Johnson
- Department of Pathology, University of Washington Seattle, WA, USA ; Department of Genetics, Albert Einstein College of Medicine New York, NY, USA
| | - Melana E Yanos
- Department of Pathology, University of Washington Seattle, WA, USA ; Department of Psychology, University of Washington Seattle, WA, USA
| | - Alessandro Bitto
- Department of Pathology, University of Washington Seattle, WA, USA
| | - Anthony Castanza
- Department of Pathology, University of Washington Seattle, WA, USA
| | - Arni Gagnidze
- Department of Pathology, University of Washington Seattle, WA, USA
| | - Brenda Gonzalez
- Department of Genetics, Albert Einstein College of Medicine New York, NY, USA
| | - Kanav Gupta
- Department of Pathology, University of Washington Seattle, WA, USA
| | - Jessica Hui
- Department of Pathology, University of Washington Seattle, WA, USA
| | - Conner Jarvie
- Department of Pathology, University of Washington Seattle, WA, USA
| | | | - Nicolas Letexier
- Department of Pathology, University of Washington Seattle, WA, USA
| | - Lanny McCanta
- Department of Pathology, University of Washington Seattle, WA, USA
| | - Maya Sangesland
- Department of Pathology, University of Washington Seattle, WA, USA
| | - Oliver Tamis
- Department of Pathology, University of Washington Seattle, WA, USA
| | - Lauren Uhde
- Department of Pathology, University of Washington Seattle, WA, USA
| | | | | | - Yousin Suh
- Department of Genetics, Albert Einstein College of Medicine New York, NY, USA
| | - Matt Kaeberlein
- Department of Pathology, University of Washington Seattle, WA, USA
| |
Collapse
|
34
|
Roforth MM, Farr JN, Fujita K, McCready LK, Atkinson EJ, Therneau TM, Cunningham JM, Drake MT, Monroe DG, Khosla S. Global transcriptional profiling using RNA sequencing and DNA methylation patterns in highly enriched mesenchymal cells from young versus elderly women. Bone 2015; 76:49-57. [PMID: 25827254 PMCID: PMC4447531 DOI: 10.1016/j.bone.2015.03.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 02/06/2015] [Accepted: 03/20/2015] [Indexed: 12/20/2022]
Abstract
Age-related bone loss in humans is associated with a decrease in bone formation relative to bone resorption, although the mechanisms for this impairment in bone formation with aging are not well understood. It is known that the precursors for the bone-forming osteoblasts reside in the mesenchymal cell population in bone marrow. Thus, in an effort to identify relevant genetic pathways that are altered with aging, we examined the gene expression and DNA methylation patterns from a highly enriched bone marrow mesenchymal cell population from young (mean age, 28.7 years) versus old (mean age, 73.3 years) women. Bone marrow mononuclear cells from these women were depleted of hematopoietic lineage (lin) and endothelial cells using a combination of magnetic- and fluorescence-activated cell sorting, yielding a previously characterized mesenchymal cell population (lin-/CD34-/CD31- cells) that is capable of osteoblast differentiation. Whole transcriptome RNA sequencing (RNAseq) of freshly isolated cells (without in vitro culture) identified 279 differentially expressed genes (p < 0.05, false discovery rate [q]< 0.10) between the young and old subjects. Pathway analysis revealed statistically significant (all p < 0.05) alterations in protein synthesis and degradation pathways, as well as mTOR, gap junction, calcium, melatonin and NFAT signaling pathways. Further, Reduced Representational Bisulphite sequencing (RRBS DNA methylation sequencing) revealed significant differences in methylation between the young and old subjects surrounding the promoters of 1528 target genes that also exhibited significant differences in gene expression by RNAseq. In summary, these studies provide novel insights into potential pathways affected by aging in a highly enriched human mesenchymal cell population analyzed without the confounding effects of in vitro culture. Specifically, our finding of alterations in several genes and pathways leading to impaired protein synthesis and turnover with aging in bone marrow mesenchymal cells points to the need for further studies examining how these changes, as well as the other alterations with aging that we identified, may contribute to the age-related impairment in osteoblast formation and/or function.
Collapse
Affiliation(s)
- Matthew M Roforth
- Endocrine Research Unit and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| | - Joshua N Farr
- Endocrine Research Unit and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| | - Koji Fujita
- Endocrine Research Unit and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| | - Louise K McCready
- Endocrine Research Unit and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| | | | | | - Julie M Cunningham
- Experimental Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Matthew T Drake
- Endocrine Research Unit and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| | - David G Monroe
- Endocrine Research Unit and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| | - Sundeep Khosla
- Endocrine Research Unit and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
35
|
Kaeberlein M. Rapamycin and ageing: when, for how long, and how much? J Genet Genomics 2014; 41:459-63. [PMID: 25269671 PMCID: PMC4401992 DOI: 10.1016/j.jgg.2014.06.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 06/18/2014] [Accepted: 06/28/2014] [Indexed: 12/31/2022]
Abstract
The drug rapamycin is the only pharmacological agent thus far shown to reproducibly extend lifespan and delay a subset of age-associated pathologies in multiple strains of mice. Unfortunately, the vast majority of aging-related studies on rapamycin in mice have been performed at a single dose of the drug delivered in encapsulated form through the diet. Recently, the National Institute on Aging Interventions Testing Program reported that a three-fold higher dose of dietary rapamycin results in a significantly greater increase in lifespan. This observation demonstrates that current studies of the effects of rapamycin on lifespan and healthspan in mice are being performed under conditions that are sub-optimal. Here I argue that the failure to properly determine the dose and timing response profile for rapamycin with respect to healthy aging represents a major barrier for the field. This barrier continues to hamper our ability to gain mechanistic insights and may threaten efforts to translate these findings into interventions that promote healthy aging in people.
Collapse
Affiliation(s)
- Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, WA 98195-7470, USA.
| |
Collapse
|
36
|
Mechanisms underlying the anti-aging and anti-tumor effects of lithocholic bile acid. Int J Mol Sci 2014; 15:16522-43. [PMID: 25238416 PMCID: PMC4200844 DOI: 10.3390/ijms150916522] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 08/21/2014] [Accepted: 09/11/2014] [Indexed: 12/13/2022] Open
Abstract
Bile acids are cholesterol-derived bioactive lipids that play essential roles in the maintenance of a heathy lifespan. These amphipathic molecules with detergent-like properties display numerous beneficial effects on various longevity- and healthspan-promoting processes in evolutionarily distant organisms. Recent studies revealed that lithocholic bile acid not only causes a considerable lifespan extension in yeast, but also exhibits a substantial cytotoxic effect in cultured cancer cells derived from different tissues and organisms. The molecular and cellular mechanisms underlying the robust anti-aging and anti-tumor effects of lithocholic acid have emerged. This review summarizes the current knowledge of these mechanisms, outlines the most important unanswered questions and suggests directions for future research.
Collapse
|
37
|
Harputlugil E, Hine C, Vargas D, Robertson L, Manning BD, Mitchell JR. The TSC complex is required for the benefits of dietary protein restriction on stress resistance in vivo. Cell Rep 2014; 8:1160-70. [PMID: 25131199 PMCID: PMC4260622 DOI: 10.1016/j.celrep.2014.07.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 06/10/2014] [Accepted: 07/14/2014] [Indexed: 02/07/2023] Open
Abstract
Protein restriction (PR) is important for the benefits of dietary restriction on longevity and stress resistance, but relevant nutrient sensors and downstream effectors in mammals remain poorly defined. We used PR-mediated protection from hepatic ischemia reperfusion injury to probe genetic requirements for the evolutionarily conserved nutrient sensors GCN2 and mTORC1 in stress resistance. One week of PR reduced free amino acids and circulating growth factors, activating GCN2 and mTORC1 repressor tuberous sclerosis complex (TSC). However, although GCN2 was dispensable for PR-induced protection, hepatic TSC1 was required. PR improved hepatic insulin sensitivity in a TSC1-dependent manner prior to ischemia, facilitating increased prosurvival signaling and reduced apoptosis after reperfusion. These benefits were partially abrogated by pharmacological PI3K inhibition or genetic deletion of the insulin receptor in hepatocytes. In conclusion, improved insulin sensitivity upon short-term PR required TSC1, facilitated increased prosurvival signaling after injury, and contributed partially to PR-mediated resistance to clinically relevant ischemia reperfusion injury.
Collapse
Affiliation(s)
- Eylul Harputlugil
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Christopher Hine
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Dorathy Vargas
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Lauren Robertson
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Brendan D Manning
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - James R Mitchell
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA.
| |
Collapse
|
38
|
Zhang J, Liu F. Tissue-specific insulin signaling in the regulation of metabolism and aging. IUBMB Life 2014; 66:485-95. [PMID: 25087968 DOI: 10.1002/iub.1293] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 07/14/2014] [Indexed: 12/30/2022]
Abstract
In mammals, insulin signaling regulates glucose homeostasis and plays an essential role in metabolism, organ growth, development, fertility, and lifespan. The defects in this signaling pathway contribute to various metabolic diseases such as type 2 diabetes, polycystic ovarian disease, hypertension, hyperlipidemia, and atherosclerosis. However, reducing the insulin signaling pathway has been found to increase longevity and delay the aging-associated diseases in various animals, ranging from nematodes to mice. These seemly paradoxical findings raise an interesting question as to how modulation of the insulin signaling pathway could be an effective approach to improve metabolism and aging. In this review, we summarize current understanding on tissue-specific functions of insulin signaling in the regulation of metabolism and lifespan. We also discuss the potential benefits and limitations in modulating tissue-specific insulin signaling pathway to improve metabolism and healthspan.
Collapse
Affiliation(s)
- Jingjing Zhang
- Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education; Diabetes Center, Institute of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | | |
Collapse
|
39
|
Abstract
Here we discuss the latest progress in development of some kinase inhibitors such as inhibitors of c-MET, LIM and Bcr-Abl kinases. Importantly, many oncogenic kinases signal via the mTOR pathway, suggesting a common target for drug combinations.
Collapse
|
40
|
Popovich IG, Anisimov VN, Zabezhinski MA, Semenchenko AV, Tyndyk ML, Yurova MN, Blagosklonny MV. Lifespan extension and cancer prevention in HER-2/neu transgenic mice treated with low intermittent doses of rapamycin. Cancer Biol Ther 2014; 15:586-92. [PMID: 24556924 DOI: 10.4161/cbt.28164] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Target of Rapamycin (TOR) is involved in cellular and organismal aging. Rapamycin extends lifespan and delays cancer in mice. It is important to determine the minimum effective dose and frequency of its administration that still extends lifespan and prevents cancer. Previously we tested 1.5 mg/kg of rapamycin given subcutaneously 6 times per two weeks followed by a two-week break (1.5 × 6/bi-weekly schedule: total of 6 injections during a 4-week period). This intermittent treatment prolonged lifespan and delayed cancer in cancer-prone female FVB/N HER-2/neu mice. Here, the dose was decreased from 1.5 mg/kg to 0.45 mg/kg per injection. This treatment was started at the age of 2 months (group Rap-2), 4 months (Rap-4), and 5 months (Rap-5). Three control groups received the solvent from the same ages. Rapamycin significantly delayed cancer and decreased tumor burden in Rap-2 and Rap-5 groups, increased mean lifespan in Rap-4 and Rap-5 groups, and increased maximal lifespan in Rap-2 and Rap-5 groups. In Rap-4 group, mean lifespan extension was achieved without significant cancer prevention. The complex relationship between life-extension and cancer-prevention depends on both the direct effect of rapamycin on cancer cells and its anti-aging effect on the organism, which in turn prevents cancer indirectly. We conclude that total doses of rapamycin that are an order of magnitude lower than standard total doses can detectably extend life span in cancer-prone mice.
Collapse
Affiliation(s)
- Irina G Popovich
- Department of Carcinogenesis and Oncogerontology; N.N. Petrov Research Institute of Oncology; St. Petersburg, Russia
| | - Vladimir N Anisimov
- Department of Carcinogenesis and Oncogerontology; N.N. Petrov Research Institute of Oncology; St. Petersburg, Russia
| | - Mark A Zabezhinski
- Department of Carcinogenesis and Oncogerontology; N.N. Petrov Research Institute of Oncology; St. Petersburg, Russia
| | - Anna V Semenchenko
- Department of Carcinogenesis and Oncogerontology; N.N. Petrov Research Institute of Oncology; St. Petersburg, Russia
| | - Margarita L Tyndyk
- Department of Carcinogenesis and Oncogerontology; N.N. Petrov Research Institute of Oncology; St. Petersburg, Russia
| | - Maria N Yurova
- Department of Carcinogenesis and Oncogerontology; N.N. Petrov Research Institute of Oncology; St. Petersburg, Russia
| | | |
Collapse
|