1
|
Giommetti A, Papanikolaou E. Advancements in Hematopoietic Stem Cell Gene Therapy: A Journey of Progress for Viral Transduction. Cells 2024; 13:1039. [PMID: 38920667 PMCID: PMC11201829 DOI: 10.3390/cells13121039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Hematopoietic stem cell (HSC) transduction has undergone remarkable advancements in recent years, revolutionizing the landscape of gene therapy specifically for inherited hematologic disorders. The evolution of viral vector-based transduction technologies, including retroviral and lentiviral vectors, has significantly enhanced the efficiency and specificity of gene delivery to HSCs. Additionally, the emergence of small molecules acting as transduction enhancers has addressed critical barriers in HSC transduction, unlocking new possibilities for therapeutic intervention. Furthermore, the advent of gene editing technologies, notably CRISPR-Cas9, has empowered precise genome modification in HSCs, paving the way for targeted gene correction. These striking progresses have led to the clinical approval of medicinal products based on engineered HSCs with impressive therapeutic benefits for patients. This review provides a comprehensive overview of the collective progress in HSC transduction via viral vectors for gene therapy with a specific focus on transduction enhancers, highlighting the latest key developments, challenges, and future directions towards personalized and curative treatments.
Collapse
Affiliation(s)
- Aurora Giommetti
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany;
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Eleni Papanikolaou
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany;
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| |
Collapse
|
2
|
Zhou H, Li I, Bramlett CS, Wang B, Hao J, Yen DP, Ando Y, Fraser SE, Lu R, Shen K. Label-free metabolic optical biomarkers track stem cell fate transition in real time. SCIENCE ADVANCES 2024; 10:eadi6770. [PMID: 38718114 PMCID: PMC11078180 DOI: 10.1126/sciadv.adi6770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 04/04/2024] [Indexed: 05/12/2024]
Abstract
Tracking stem cell fate transition is crucial for understanding their development and optimizing biomanufacturing. Destructive single-cell methods provide a pseudotemporal landscape of stem cell differentiation but cannot monitor stem cell fate in real time. We established a metabolic optical metric using label-free fluorescence lifetime imaging microscopy (FLIM), feature extraction and machine learning-assisted analysis, for real-time cell fate tracking. From a library of 205 metabolic optical biomarker (MOB) features, we identified 56 associated with hematopoietic stem cell (HSC) differentiation. These features collectively describe HSC fate transition and detect its bifurcate lineage choice. We further derived a MOB score measuring the "metabolic stemness" of single cells and distinguishing their division patterns. This score reveals a distinct role of asymmetric division in rescuing stem cells with compromised metabolic stemness and a unique mechanism of PI3K inhibition in promoting ex vivo HSC maintenance. MOB profiling is a powerful tool for tracking stem cell fate transition and improving their biomanufacturing from a single-cell perspective.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Irene Li
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Charles S. Bramlett
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Bowen Wang
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jia Hao
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Daniel P. Yen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Yuta Ando
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Scott E. Fraser
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA
- Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Rong Lu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
- Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
- USC Stem Cell, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
3
|
Ji S, Xiong M, Chen H, Liu Y, Zhou L, Hong Y, Wang M, Wang C, Fu X, Sun X. Cellular rejuvenation: molecular mechanisms and potential therapeutic interventions for diseases. Signal Transduct Target Ther 2023; 8:116. [PMID: 36918530 PMCID: PMC10015098 DOI: 10.1038/s41392-023-01343-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/16/2022] [Accepted: 01/19/2023] [Indexed: 03/16/2023] Open
Abstract
The ageing process is a systemic decline from cellular dysfunction to organ degeneration, with more predisposition to deteriorated disorders. Rejuvenation refers to giving aged cells or organisms more youthful characteristics through various techniques, such as cellular reprogramming and epigenetic regulation. The great leaps in cellular rejuvenation prove that ageing is not a one-way street, and many rejuvenative interventions have emerged to delay and even reverse the ageing process. Defining the mechanism by which roadblocks and signaling inputs influence complex ageing programs is essential for understanding and developing rejuvenative strategies. Here, we discuss the intrinsic and extrinsic factors that counteract cell rejuvenation, and the targeted cells and core mechanisms involved in this process. Then, we critically summarize the latest advances in state-of-art strategies of cellular rejuvenation. Various rejuvenation methods also provide insights for treating specific ageing-related diseases, including cellular reprogramming, the removal of senescence cells (SCs) and suppression of senescence-associated secretory phenotype (SASP), metabolic manipulation, stem cells-associated therapy, dietary restriction, immune rejuvenation and heterochronic transplantation, etc. The potential applications of rejuvenation therapy also extend to cancer treatment. Finally, we analyze in detail the therapeutic opportunities and challenges of rejuvenation technology. Deciphering rejuvenation interventions will provide further insights into anti-ageing and ageing-related disease treatment in clinical settings.
Collapse
Affiliation(s)
- Shuaifei Ji
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Mingchen Xiong
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Huating Chen
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Yiqiong Liu
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Laixian Zhou
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Yiyue Hong
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Mengyang Wang
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, 999078, Macau SAR, China.
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China.
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China.
| |
Collapse
|
4
|
Blagosklonny MV. Cellular senescence: when growth stimulation meets cell cycle arrest. Aging (Albany NY) 2023; 15:905-913. [PMID: 36805938 PMCID: PMC10008486 DOI: 10.18632/aging.204543] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/16/2023] [Indexed: 02/21/2023]
Abstract
At the very moment of cell-cycle arrest, the cell is not senescent yet. For several days in cell culture, the arrested cell is acquiring a senescent phenotype. What is happening during this geroconversion? Cellular enlargement (hypertrophy) and hyperfunctions (lysosomal and hyper-secretory) are hallmarks of geroconversion.
Collapse
|
5
|
Arif T. Lysosomes and Their Role in Regulating the Metabolism of Hematopoietic Stem Cells. BIOLOGY 2022; 11:1410. [PMID: 36290314 PMCID: PMC9598322 DOI: 10.3390/biology11101410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/26/2022]
Abstract
Hematopoietic stem cells (HSCs) have the capacity to renew blood cells at all stages of life and are largely quiescent at a steady state. It is essential to understand the processes that govern quiescence in HSCs to enhance bone marrow transplantation. It is hypothesized that in their quiescent state, HSCs primarily use glycolysis for energy production rather than mitochondrial oxidative phosphorylation (OXPHOS). In addition, the HSC switch from quiescence to activation occurs along a continuous developmental path that is driven by metabolism. Specifying the metabolic regulation pathway of HSC quiescence will provide insights into HSC homeostasis for therapeutic application. Therefore, understanding the metabolic demands of HSCs at a steady state is key to developing innovative hematological therapeutics. Lysosomes are the major degradative organelle in eukaryotic cells. Catabolic, anabolic, and lysosomal function abnormalities are connected to an expanding list of diseases. In recent years, lysosomes have emerged as control centers of cellular metabolism, particularly in HSC quiescence, and essential regulators of cell signaling have been found on the lysosomal membrane. In addition to autophagic processes, lysosomal activities have been shown to be crucial in sustaining quiescence by restricting HSCs access to a nutritional reserve essential for their activation into the cell cycle. Lysosomal activity may preserve HSC quiescence by altering glycolysis-mitochondrial biogenesis. The understanding of HSC metabolism has significantly expanded over the decade, revealing previously unknown requirements of HSCs in both their dividing (active) and quiescent states. Therefore, understanding the role of lysosomes in HSCs will allow for the development of innovative treatment methods based on HSCs to fight clonal hematopoiesis and HSC aging.
Collapse
Affiliation(s)
- Tasleem Arif
- Department of Cell, Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
6
|
Keyvani Chahi A, Belew MS, Xu J, Chen HTT, Rentas S, Voisin V, Krivdova G, Lechman E, Marhon SA, De Carvalho DD, Dick JE, Bader GD, Hope KJ. PLAG1 dampens protein synthesis to promote human hematopoietic stem cell self-renewal. Blood 2022; 140:992-1008. [PMID: 35639948 PMCID: PMC9437713 DOI: 10.1182/blood.2021014698] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 05/12/2022] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic stem cell (HSC) dormancy is understood as supportive of HSC function and its long-term integrity. Although regulation of stress responses incurred as a result of HSC activation is recognized as important in maintaining stem cell function, little is understood of the preventive machinery present in human HSCs that may serve to resist their activation and promote HSC self-renewal. We demonstrate that the transcription factor PLAG1 is essential for long-term HSC function and, when overexpressed, endows a 15.6-fold enhancement in the frequency of functional HSCs in stimulatory conditions. Genome-wide measures of chromatin occupancy and PLAG1-directed gene expression changes combined with functional measures reveal that PLAG1 dampens protein synthesis, restrains cell growth and division, and enhances survival, with the primitive cell advantages it imparts being attenuated by addition of the potent translation activator, c-MYC. We find PLAG1 capitalizes on multiple regulatory factors to ensure protective diminished protein synthesis including 4EBP1 and translation-targeting miR-127 and does so independently of stress response signaling. Overall, our study identifies PLAG1 as an enforcer of human HSC dormancy and self-renewal through its highly context-specific regulation of protein biosynthesis and classifies PLAG1 among a rare set of bona fide regulators of messenger RNA translation in these cells. Our findings showcase the importance of regulated translation control underlying human HSC physiology, its dysregulation under activating demands, and the potential if its targeting for therapeutic benefit.
Collapse
Affiliation(s)
- Ava Keyvani Chahi
- Department of Biochemistry and Biomedical Sciences,McMaster University, Hamilton, ON, Canada
| | - Muluken S Belew
- Department of Biochemistry and Biomedical Sciences,McMaster University, Hamilton, ON, Canada
| | - Joshua Xu
- Department of Biochemistry and Biomedical Sciences,McMaster University, Hamilton, ON, Canada
| | - He Tian Tony Chen
- Department of Biochemistry and Biomedical Sciences,McMaster University, Hamilton, ON, Canada
| | - Stefan Rentas
- Department of Biochemistry and Biomedical Sciences,McMaster University, Hamilton, ON, Canada
| | | | - Gabriela Krivdova
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Eric Lechman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; and
| | - Sajid A Marhon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; and
| | - Daniel D De Carvalho
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; and
- Department of Medical Biophysics and
| | - John E Dick
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; and
| | - Gary D Bader
- The Donnelly Centre and
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; and
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Kristin J Hope
- Department of Biochemistry and Biomedical Sciences,McMaster University, Hamilton, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; and
- Department of Medical Biophysics and
| |
Collapse
|
7
|
Xuan J, Liu Y, Liu J, Zeng X, Wang H. New Insights into Hematopoietic Stem Cell Expansion to Stimulate Repopulation of the Adult Blood System for Transplantation. Life (Basel) 2022; 12:life12050716. [PMID: 35629383 PMCID: PMC9146250 DOI: 10.3390/life12050716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
Successful engraftment of hematopoietic stem cells (HSCs) and progenitor cells (HSPCs) may be considered as a basis for the repopulation of the blood cells after transplantation in adults. Therefore, in vivo and ex vivo expansion of HSCs holds great promise for clinical applications. In this review, the mechanisms of HSC expansion will be discussed, considering the previous studies and works of literature. This is aimed to identify the signaling pathways that regulate HSC expansion and improve the application of engraftment in disease management. The following aspects will be included: (i) Stimulation of HSCs growth in vivo through gene regulation and cytokines activation; (ii) direct or indirect induction of HSC expansion by regulating signaling pathways; (iii) addition to assisting cells to help in the proliferation of HSCs; (iv) changing of living environment in the HSCs cultures via adjusting components and forms of cultures; (v) enhancement of HSC expansion by incorporating substances, such as extracellular vesicles (EVs), UM171, among others. In this review, recent new findings that provide us with new insights into HSC expansion methods have been summarized. Furthermore, these findings will also provide more possibilities for the development of some novel strategies for expanding and engrafting HSCs applied for treatments of some hematopoietic disorders.
Collapse
Affiliation(s)
- Jiangying Xuan
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (J.L.); (X.Z.)
- Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Yingxia Liu
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (J.L.); (X.Z.)
| | - Jinhui Liu
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (J.L.); (X.Z.)
| | - Xiaoping Zeng
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (J.L.); (X.Z.)
| | - Hongmei Wang
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (J.L.); (X.Z.)
- Correspondence: ; Tel.: +86-137-6700-4966
| |
Collapse
|
8
|
Abstract
A hallmark of cellular senescence is proliferation-like activity of growth-promoting pathways (such as mTOR and MAPK) in non-proliferating cells. When the cell cycle is arrested, these pathways convert arrest to senescence (geroconversion), rendering cells hypertrophic, beta-Gal-positive and hyperfunctional. The senescence-associated secretory phenotype (SASP) is one of the numerous hyperfunctions. Figuratively, geroconversion is a continuation of growth in non-proliferating cells. Rapamycin, a reversible inhibitor of growth, slows down mTOR-driven geroconversion. Developed two decades ago, this model had accurately predicted that rapamycin must extend life span of animals. However, the notion that senescent cells directly cause organismal aging is oversimplified. Senescent cells contribute to organismal aging but are not strictly required. Cell senescence and organismal aging can be linked indirectly via the same underlying cause, namely hyperfunctional signaling pathways such as mTOR.
Collapse
|
9
|
Li Q, Hao S, Cheng T. [Research progress on in vitro expansion and clinical application of hematopoietic stem cell]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2022; 43:167-172. [PMID: 35381684 PMCID: PMC8980649 DOI: 10.3760/cma.j.issn.0253-2727.2022.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Q Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - S Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - T Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
10
|
Tadokoro Y, Hirao A. The Role of Nutrients in Maintaining Hematopoietic Stem Cells and Healthy Hematopoiesis for Life. Int J Mol Sci 2022; 23:1574. [PMID: 35163498 PMCID: PMC8836201 DOI: 10.3390/ijms23031574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/26/2022] [Accepted: 01/26/2022] [Indexed: 02/04/2023] Open
Abstract
Nutrients are converted by the body to smaller molecules, which are utilized for both anabolic and catabolic metabolic reactions. Cooperative regulation of these processes is critical for life-sustaining activities. In this review, we focus on how the regulation of nutrient-driven metabolism maintains healthy hematopoietic stem cells (HSCs). For this purpose, we have examined the metabolic regulation of HSCs from two perspectives: (1) the control of intracellular metabolism by the balance of anabolic and catabolic reactions; and (2) the control of organismal metabolic status and hematopoiesis by dietary intake of nutrients. Critical roles of catabolic regulators in stem cell homeostasis are conserved in several types of tissues, including hematopoiesis. These catabolic signals are also major regulators of organismal lifespan in multiple species. In parallel, changes to nutrients via alterations to dietary intake affect not only an organism's metabolic state but also the behavior of its stem cells. While the molecular mechanisms involved in these two aspects of nutrient function may not necessarily overlap, a deeper understanding of these phenomena will point to new avenues of medical research and may furnish new agents for improving human health care.
Collapse
Affiliation(s)
- Yuko Tadokoro
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| | - Atsushi Hirao
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| |
Collapse
|
11
|
Atkinson SP. Previews. Stem Cells 2021. [DOI: 10.1002/stem.3460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
12
|
Wu F, Chen Z, Liu J, Hou Y. The Akt-mTOR network at the interface of hematopoietic stem cell homeostasis. Exp Hematol 2021; 103:15-23. [PMID: 34464661 DOI: 10.1016/j.exphem.2021.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/15/2022]
Abstract
Hematopoietic stem cells (HSCs) are immature blood cells that exhibit multilineage differentiation capacity. Homeostasis is critical for HSC potential and lifelong hematopoiesis, and HSC homeostasis is tightly governed by both intrinsic molecular networks and microenvironmental signals. The evolutionarily conserved serine/threonine protein kinase B (PKB, also referred to as Akt)-mammalian target of rapamycin (mTOR) pathway is universal to nearly all multicellular organisms and plays an integral role in most cellular processes. Emerging evidence has revealed a central role of the Akt-mTOR network in HSC homeostasis, because it responds to multiple intracellular and extracellular signals and regulates various downstream targets, eventually affecting several cellular processes, including the cell cycle, mitochondrial metabolism, and protein synthesis. Dysregulated Akt-mTOR signaling greatly affects HSC self-renewal, maintenance, differentiation, survival, autophagy, and aging, as well as transformation of HSCs to leukemia stem cells. Here, we review recent works and provide an advanced understanding of how the Akt-mTOR network regulates HSC homeostasis, thus offering insights into future clinical applications.
Collapse
Affiliation(s)
- Feng Wu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Zhe Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jingbo Liu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China.
| | - Yu Hou
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
13
|
Anti-aging: senolytics or gerostatics (unconventional view). Oncotarget 2021; 12:1821-1835. [PMID: 34504654 PMCID: PMC8416555 DOI: 10.18632/oncotarget.28049] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/05/2021] [Indexed: 12/17/2022] Open
Abstract
Senolytics are basically anti-cancer drugs, repurposed to kill senescent cells selectively. It is even more difficult to selectively kill senescent cells than to kill cancer cells. Based on lessons of cancer therapy, here I suggest how to exploit oncogene-addiction and to combine drugs to achieve selectivity. However, even if selective senolytic combinations will be developed, there is little evidence that a few senescent cells are responsible for organismal aging. I also discuss gerostatics, such as rapamycin and other rapalogs, pan-mTOR inhibitors, dual PI3K/mTOR inhibitors, which inhibit growth- and aging-promoting pathways. Unlike senolytics, gerostatics do not kill cells but slow down cellular geroconversion to senescence. Numerous studies demonstrated that inhibition of the mTOR pathways by any means (genetic, pharmacological and dietary) extends lifespan. Currently, only two studies demonstrated that senolytics (fisetin and a combination Dasatinib plus Quercetin) extend lifespan in mice. These senolytics slightly inhibit the mTOR pathway. Thus, life extension by these senolytics can be explained by their slight rapamycin-like (gerostatic) effects.
Collapse
|
14
|
Stergiou IE, Kapsogeorgou EK. Autophagy and Metabolism in Normal and Malignant Hematopoiesis. Int J Mol Sci 2021; 22:8540. [PMID: 34445246 PMCID: PMC8395194 DOI: 10.3390/ijms22168540] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
The hematopoietic system relies on regulation of both metabolism and autophagy to maintain its homeostasis, ensuring the self-renewal and multipotent differentiation potential of hematopoietic stem cells (HSCs). HSCs display a distinct metabolic profile from that of their differentiated progeny, while metabolic rewiring from glycolysis to oxidative phosphorylation (OXPHOS) has been shown to be crucial for effective hematopoietic differentiation. Autophagy-mediated regulation of metabolism modulates the distinct characteristics of quiescent and differentiating hematopoietic cells. In particular, mitophagy determines the cellular mitochondrial content, thus modifying the level of OXPHOS at the different differentiation stages of hematopoietic cells, while, at the same time, it ensures the building blocks and energy for differentiation. Aberrations in both the metabolic status and regulation of the autophagic machinery are implicated in the development of hematologic malignancies, especially in leukemogenesis. In this review, we aim to investigate the role of metabolism and autophagy, as well as their interconnections, in normal and malignant hematopoiesis.
Collapse
Affiliation(s)
| | - Efstathia K. Kapsogeorgou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
15
|
Tousian H, Razavi BM, Hosseinzadeh H. In search of elixir: Pharmacological agents against stem cell senescence. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:868-880. [PMID: 34712416 PMCID: PMC8528253 DOI: 10.22038/ijbms.2021.51917.11773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 03/02/2021] [Indexed: 12/13/2022]
Abstract
Stem cell senescence causes different complications. In addition to the aging phenomenon, stem cell senescence has been investigated in various concepts such as cancer, adverse drug effects, and as a limiting factor in cell therapy. This manuscript examines protective medicines and supplements which are capable of hindering stem cell senescence. We searched the databases such as EMBASE, PubMed, and Web of Science with the keywords "stem cell," "progenitor cell," "satellite," "senescence" and excluded the keywords "cancer," "tumor," "malignancy" and "carcinoma" until June 2020. Among these results, we chose 47 relevant studies. Our investigation indicates that most of these studies examined endothelial progenitor cells, hematopoietic stem cells, mesenchymal stem cells, adipose-derived stem cells, and a few others were about less-discussed types of stem cells such as cardiac stem cells, myeloblasts, and induced pluripotent stem cells. From another aspect, 17β-Estradiol, melatonin, metformin, rapamycin, coenzyme Q10, N-acetyl cysteine, and vitamin C were the most studied agents, while the main protective mechanism was through telomerase activity enhancement or oxidative damage ablation. Although many of these studies are in vitro, they are still worthwhile. Stem cell senescence in the in vitro expansion stage is an essential concern in clinical procedures of cell therapy. Moreover, in vitro studies are the first step for further in vivo and clinical studies. It is noteworthy to mention the fact that these protective agents have been used in the clinical setting for various purposes for a long time. Given that, we only need to examine their systemic anti-senescence effects and effective dosages.
Collapse
Affiliation(s)
- Hourieh Tousian
- Vice-chancellery of Food and Drug,Shahroud University of Medical Sciences, Shahroud, Iran
| | - Bibi Marjan Razavi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
Lin RJ, Elias HK, van den Brink MRM. Immune Reconstitution in the Aging Host: Opportunities for Mechanism-Based Therapy in Allogeneic Hematopoietic Cell Transplantation. Front Immunol 2021; 12:674093. [PMID: 33953731 PMCID: PMC8089387 DOI: 10.3389/fimmu.2021.674093] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 03/30/2021] [Indexed: 12/13/2022] Open
Abstract
Older patients with hematologic malignancies are increasingly considered for allogeneic hematopoietic cell transplantation with encouraging outcomes. While aging-related thymic dysfunction remains a major obstacle to optimal and timely immune reconstitution post- transplantation, recent accumulating evidence has suggested that various aging hallmarks such as cellular senescence, inflamm-aging, and hematopoietic stem cell exhaustion, could also impact immune reconstitution post-transplantation in both thymic-dependent and independent manner. Here we review molecular and cellular aspects of immune senescence and immune rejuvenation related to allogeneic hematopoietic cell transplantation among older patients and discuss potential strategies for mechanism-based therapeutic intervention.
Collapse
Affiliation(s)
- Richard J Lin
- Adult Bone Marrow Transplantation (BMT) Service, Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Harold K Elias
- Adult Bone Marrow Transplantation (BMT) Service, Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation (BMT) Service, Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
17
|
Abstract
Blood is generated throughout life by continued proliferation and differentiation of hematopoietic progenitors, while at the top of the hierarchy, hematopoietic stem cells (HSCs) remain largely quiescent. This way HSCs avoid senescence and preserve their capacity to repopulate the hematopoietic system. But HSCs are not always quiescent, proliferating extensively in conditions such as those found in the fetal liver. Understanding the elusive mechanisms that regulate HSC fate would enable us to comprehend a crucial piece of HSC biology and pave the way for ex-vivo HSC expansion with clear clinical benefit. Here we review how metabolism, endoplasmic reticulum stress and oxidative stress condition impact HSCs decision to self-renew or differentiate and how these signals integrate into the mammalian target of rapamycin (mTOR) pathway. We argue that the bone marrow microenvironment continuously favors differentiation through the activation of the mTOR complex (mTORC)1 signaling, while the fetal liver microenvironment favors self-renewal through the inverse mechanism. In addition, we also postulate that strategies that have successfully achieved HSC expansion, directly or indirectly, lead to the inactivation of mTORC1. Finally, we propose a mechanism by which mTOR signaling, during cell division, conditions HSC fate. This mechanism has already been demonstrated in mature hematopoietic cells (T-cells), that face a similar decision after activation, either undergoing clonal expansion or differentiation.
Collapse
|
18
|
Lin Z, Hollinger MK, Wu Z, Sun W, Batey K, Kim J, Chen J, Feng X, Young NS. Sirolimus augments hematopoietic stem and progenitor cell regeneration following hematopoietic insults. Stem Cells 2021; 39:240-252. [PMID: 33270949 PMCID: PMC7898520 DOI: 10.1002/stem.3313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 11/17/2020] [Indexed: 04/20/2023]
Abstract
The role of mammalian target of rapamycin and its suppressor sirolimus in the regulation of hematopoietic stem and progenitor cells (HSPCs) is controversial. We show here that sirolimus enhanced regeneration of HSPCs in mice exposed to sublethal total body irradiation (TBI) and other regenerative stressors. Sorted Lin- CD150+ bone marrow cells from sirolimus-treated TBI mice had increased expression of c-Kit and other hematopoietic genes. HSPCs from sirolimus-treated TBI mice were functionally competent when tested by competitive engraftment in vivo. Postradiation regeneration of HSPCs in mice treated with sirolimus was accompanied by decreased γ-H2AX levels detected by flow cytometry and increased expression of DNA repair genes by quantitative polymerase chain reaction. Reduction of cell death and DNA damage post-radiation by sirolimus was associated with enhanced clearance of cellular reactive oxygen species (ROS) in HSPCs. Increased HSPC recovery with sirolimus was also observed in mice injected with hematoxic agents, busulfan and 5-fluorouracil. In contrast, sirolimus showed no effect on HSPCs in normal mice at steady state, but stimulated HSPC expansion in mice carrying the Wv mutation at the c-Kit locus. In human to mouse xenotransplantation, sirolimus enhanced engraftment of irradiated human CD34+ cells. In summary, our results are consistent with sirolimus' acceleration of HSPC recovery in response to hematopoietic stress, associated with reduced DNA damage and ROS. Sirolimus might have clinical application for the treatment and prevention of hematopoietic injury.
Collapse
Affiliation(s)
- Zenghua Lin
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMarylandUSA
- Department of HematologyAffiliated Hospital of Nantong UniversityNantongJiangsuPeople's Republic of China
| | - Maile K. Hollinger
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Zhijie Wu
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Wanling Sun
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMarylandUSA
- Department of HematologyXuanwu Hospital, Capital Medical UniversityBeijingPeople's Republic of China
| | - Kaylind Batey
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Jisoo Kim
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Jichun Chen
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Xingmin Feng
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Neal S. Young
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
19
|
Liao C, Xiao Y, Liu L. The Dynamic Process and Its Dual Effects on Tumors of Therapy-Induced Senescence. Cancer Manag Res 2021; 12:13553-13566. [PMID: 33408525 PMCID: PMC7781229 DOI: 10.2147/cmar.s285083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Cellular senescence is traditionally considered as stable cell cycle arrest state with other phenotypic alterations including the production of an array of cytokines and growth factors. Cancer cells undergo senescence in response to chemotherapeutic agents, radiotherapy and molecular targeted therapy. This form of senescence is termed therapy-induced senescence (TIS) and represents a desirable target in cancer therapy. Recent studies have shown that cellular senescence is a highly heterogeneous and dynamic process. Apart from being cleared by the immune system, the senescent cancer cells may survive for a long time and escape from senescence state. Notably, these cells even have the potential to regain stem-like state with high aggressiveness that eventually facilitates cancer recurrence. Furthermore, the senescence-associated secretory phenotype (SASP) of senescent cells is not always the same, and could establish immunosuppression and a protumor microenvironment. Given these detrimental effects, senescence-inducing chemotherapy followed by senotherapy (the “one–two punch” approach), has emerged. This combined therapy could mitigate unnecessary side effects of the persistent senescent cells, reduce the toxicity of pro-senescence therapy and prolong the survival of cancer patients, and it has a potential future in the precise treatment of cancer. Herein, we review the complex effects of therapy-induced senescence in cancer and highlight the great promise of two-step strategies in anticancer therapies.
Collapse
Affiliation(s)
- Chenxi Liao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Yin Xiao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Lingbo Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| |
Collapse
|
20
|
Chen YY, Liu YF, Liu YD, Deng XH, Zhou J. IRF7 suppresses hematopoietic regeneration under stress via CXCR4. STEM CELLS (DAYTON, OHIO) 2020; 39:183-195. [PMID: 33252829 DOI: 10.1002/stem.3308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 11/08/2020] [Indexed: 11/06/2022]
Abstract
Hematopoietic stem cells (HSCs) maintain quiescence under steady state; however, they are compelled to proliferate and expand to replenish the blood system under stress. The molecular basis underlying stress hematopoiesis remains to be fully understood. In this study, we reported that IRF7 represents an important regulator of stress hematopoiesis. Interferon regulatory factor 7 (IRF7) was dispensable for normal hematopoiesis, whereas its deficiency significantly enhanced hematopoietic stem and progenitor cells (HSPCs) regeneration and improved long-term repopulation of HSCs under stress. Mechanistic studies showed that CXCR4 was identified as a downstream target of IRF7. Overexpression of CXCR4 abrogated the enhanced proliferation and regeneration of IRF7-deficient HSPCs under stress. Similar results were obtained in HSCs from human umbilical cord blood. These observations demonstrated that IRF7 plays an important role in hematopoietic regeneration under stress.
Collapse
Affiliation(s)
- Ying-Ying Chen
- Joint Program in Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China.,Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China.,Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Yu-Feng Liu
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Yong-Dong Liu
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiao-Hui Deng
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China.,Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Jie Zhou
- Joint Program in Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China.,Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China.,Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
21
|
Jiang Y, Xu Z, Ma N, Yin L, Hao C, Li J. Effects of signaling pathway inhibitors on hematopoietic stem cells. Mol Med Rep 2020; 23:9. [PMID: 33179097 PMCID: PMC7687261 DOI: 10.3892/mmr.2020.11647] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 02/17/2020] [Indexed: 12/23/2022] Open
Abstract
While there are numerous small molecule inhibitory drugs available for a wide range of signalling pathways, at present, they are generally not used in combination in clinical settings. Previous reports have reported that the effects of glycogen synthase kinase (GSK)3β, p38MAPK, mTOR and histone deacetylase signaling combined together to suppress the stem-like nature of hematopoietic stem cells (HSCs), driving these cells to differentiate, cease proliferating and thereby impairing normal hematopoietic functionality. The present study aimed to determine the effect of HDACs, mTOR, GSK-3β and p38MAPK inhibitor combinations on the efficient expansion of HSCs using flow cytometry. Moreover, it specifically aimed to determine how inhibitors of the GSK3β signaling pathway, in combination with inhibitors of P38MAPK and mTOR signaling or histone deacetylase (HDAC) inhibitors, could affect HSC expansion, with the goal of identifying novel combination strategies useful for the expansion of HSCs. The results indicated that p38MAPK and/or GSK3β inhibitors increased Lin− cell and Lin−Sca-1+c-kit+ (LSK) cell numbers in vitro. Taken together, these results suggested that a combination of p38MAPK and GSK3β signaling may regulate HSC differentiation in vitro. These findings further indicated that the suppression of p38MAPK and/or GSK3β signalling may modulate HSC differentiation and self-renewal to enhance HSC expansion.
Collapse
Affiliation(s)
- Yuyu Jiang
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Zhaofeng Xu
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Na Ma
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Lizhi Yin
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Caiqin Hao
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Jing Li
- Stem Cell Laboratory, Department of Biology, College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| |
Collapse
|
22
|
Li X, Ma X, Chen Y, Peng D, Wang H, Chen S, Xiao Y, Li L, Zhou H, Cheng F, Gao Y, Chang J, Cheng T, Liu L. Coinhibition of activated p38 MAPKα and mTORC1 potentiates stemness maintenance of HSCs from SR1-expanded human cord blood CD34 + cells via inhibition of senescence. Stem Cells Transl Med 2020; 9:1604-1616. [PMID: 32602209 PMCID: PMC7695631 DOI: 10.1002/sctm.20-0129] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 01/10/2023] Open
Abstract
The stemness of ex vivo expanded hematopoietic stem cells (HSCs) is usually compromised by current methods. To explore the failure mechanism of stemness maintenance of human HSCs, which were expanded from human umbilical cord blood (hUCB) CD34+ cells, by differentiation inhibitor Stem Regenin 1 (SR1), an antagonist of aryl hydrocarbon receptor, we investigated the activity of p38 mitogen‐activated protein kinase α (p38 MAPKα, p38α) and mammalian target of rapamycin complex 1 (mTORC1), and their effect on SR1‐expanded hUCB CD34+ cells. Our results showed that cellular senescence occurred in the SR1‐expanded hUCB CD34+ cells in which p38α and mTORC1 were successively activated. Furthermore, their coinhibition resulted in a further decrease in hUCB CD34+ cell senescence without an effect on apoptosis, promoted the maintenance of expanded phenotypic HSCs without differentiation inhibition, increased the hematopoietic reconstitution ability of multiple lineages, and potentiated the long‐term self‐renewal capability of HSCs from SR1‐expanded hUCB CD34+ cells in NOD/Shi‐scid/IL‐2Rγnull mice. Our mechanistic study revealed that senescence inhibition by our strategy was mainly attributed to downregulation of the splicesome, proteasome formation, and pyrimidine metabolism signaling pathways. These results suggest that coinhibition of activated p38α and mTORC1 potentiates stemness maintenance of HSCs from SR1‐expanded hUCB CD34+ cells via senescence inhibition. Thus, we established a new strategy to maintain the stemness of ex vivo differentiation inhibitor‐expanded human HSCs via coinhibition of multiple independent senescence initiating signal pathways. This senescence inhibition‐induced stemness maintenance of ex vivo expanded HSCs could also have an important role in other HSC expansion systems.
Collapse
Affiliation(s)
- Xiaoyi Li
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiao Ma
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Ying Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Danyue Peng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Huifang Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Suhua Chen
- Department of Gynaecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yin Xiao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lei Li
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Hao Zhou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fanjun Cheng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Chinese Academy of Medical Science, Tianjin, People's Republic of China
| | - Jiwei Chang
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, People's Republic of China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Chinese Academy of Medical Science, Tianjin, People's Republic of China
| | - Lingbo Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
23
|
Blagosklonny MV. From causes of aging to death from COVID-19. Aging (Albany NY) 2020; 12:10004-10021. [PMID: 32534452 PMCID: PMC7346074 DOI: 10.18632/aging.103493] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022]
Abstract
COVID-19 is not deadly early in life, but mortality increases exponentially with age, which is the strongest predictor of mortality. Mortality is higher in men than in women, because men age faster, and it is especially high in patients with age-related diseases, such as diabetes and hypertension, because these diseases are manifestations of aging and a measure of biological age. At its deepest level, aging (a program-like continuation of developmental growth) is driven by inappropriately high cellular functioning. The hyperfunction theory of quasi-programmed aging explains why COVID-19 vulnerability (lethality) is an age-dependent syndrome, linking it to other age-related diseases. It also explains inflammaging and immunosenescence, hyperinflammation, hyperthrombosis, and cytokine storms, all of which are associated with COVID-19 vulnerability. Anti-aging interventions, such as rapamycin, may slow aging and age-related diseases, potentially decreasing COVID-19 vulnerability.
Collapse
|
24
|
MicroRNAs in hematopoietic stem cell aging. Mech Ageing Dev 2020; 189:111281. [PMID: 32512019 DOI: 10.1016/j.mad.2020.111281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/17/2020] [Accepted: 06/01/2020] [Indexed: 12/23/2022]
Abstract
The functional decline that is observed in HSCs upon aging is attributed mainly to cell intrinsic factors that regulate quiescence, self-renewal and differentiation. MicroRNAs (miRs) have an indispensable role in the regulation of HSCs and have been shown to also regulate processes related to tissue aging in specific cell types. Here we discuss the role of miRs in the regulation of HSC self-renewal and differentiation throughout life and its implications for future research.
Collapse
|
25
|
Tousian H, Razavi BM, Hosseinzadeh H. Looking for immortality: Review of phytotherapy for stem cell senescence. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:154-166. [PMID: 32405357 PMCID: PMC7211350 DOI: 10.22038/ijbms.2019.40223.9522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this paper, we discussed natural agents with protective effects against stem cell senescence. Different complications have been observed due to stem cell senescence and the most important of them is "Aging". Senescent cells have not normal function and their secretary inflammatory factors induce chronic inflammation in body which causes different pathologies. Stem cell senescence also has been investigated in different diseases or as drug adverse effects. We searched databases such as Embase, Pubmed and Web of Science with keywords "stem cell", "progenitor cell", "satellite", "senescence" and excluded keywords "cancer", "tumor", "malignancy" and "carcinoma" without time limitation until May 2019. Among them we chose 52 articles that have investigated protective effects of natural agents (extracts or molecules) against cellular senescence in different kind of adult stem cells. Most of these studies were in endothelial progenitor cells, hematopoietic stem cells, mesenchymal stem cells, adipose-derived stem cells and few were about other kinds of stem cells. Most studied agents were resveratrol and ginseng which are also commercially available as supplement. Most protective molecular targets were telomerase and anti-oxidant enzymes to preserve genome integrity and reduce senescence-inducing signals. Due to the safe and long history of herbal usage in clinic, phytotherapy can be used for preventing stem cell senescence and their related complication. Resveratrol and ginseng can be the first choice for this aim due to their protective mechanisms in various kinds of stem cells and their long term clinical usage.
Collapse
Affiliation(s)
- Hourieh Tousian
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Marjan Razavi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
26
|
Xie SZ, Garcia-Prat L, Voisin V, Ferrari R, Gan OI, Wagenblast E, Kaufmann KB, Zeng AGX, Takayanagi SI, Patel I, Lee EK, Jargstorf J, Holmes G, Romm G, Pan K, Shoong M, Vedi A, Luberto C, Minden MD, Bader GD, Laurenti E, Dick JE. Sphingolipid Modulation Activates Proteostasis Programs to Govern Human Hematopoietic Stem Cell Self-Renewal. Cell Stem Cell 2019; 25:639-653.e7. [PMID: 31631013 PMCID: PMC6838675 DOI: 10.1016/j.stem.2019.09.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/09/2019] [Accepted: 09/23/2019] [Indexed: 12/30/2022]
Abstract
Cellular stress responses serve as crucial decision points balancing persistence or culling of hematopoietic stem cells (HSCs) for lifelong blood production. Although strong stressors cull HSCs, the linkage between stress programs and self-renewal properties that underlie human HSC maintenance remains unknown, particularly at quiescence exit when HSCs must also dynamically shift metabolic state. Here, we demonstrate distinct wiring of the sphingolipidome across the human hematopoietic hierarchy and find that genetic or pharmacologic modulation of the sphingolipid enzyme DEGS1 regulates lineage differentiation. Inhibition of DEGS1 in hematopoietic stem and progenitor cells during the transition from quiescence to cellular activation with N-(4-hydroxyphenyl) retinamide activates coordinated stress pathways that coalesce on endoplasmic reticulum stress and autophagy programs to maintain immunophenotypic and functional HSCs. Thus, our work identifies a linkage between sphingolipid metabolism, proteostatic quality control systems, and HSC self-renewal and provides therapeutic targets for improving HSC-based cellular therapeutics.
Collapse
Affiliation(s)
- Stephanie Z Xie
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada.
| | - Laura Garcia-Prat
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada
| | - Veronique Voisin
- The Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada
| | - Robin Ferrari
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada
| | - Olga I Gan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada
| | - Elvin Wagenblast
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada
| | - Kerstin B Kaufmann
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada
| | - Andy G X Zeng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Shin-Ichiro Takayanagi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada; R&D Division, Kyowa Kirin Co., Ltd., Tokyo 194-8533, Japan
| | - Ishita Patel
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada
| | - Esther K Lee
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada
| | - Joseph Jargstorf
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada
| | - Gareth Holmes
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada
| | - Guy Romm
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada
| | - Kristele Pan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada
| | - Michelle Shoong
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada
| | - Aditi Vedi
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, UK
| | - Chiara Luberto
- Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada; Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Elisa Laurenti
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, UK
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G0A3, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S1A8, Canada.
| |
Collapse
|
27
|
Expansion of primitive human hematopoietic stem cells by culture in a zwitterionic hydrogel. Nat Med 2019; 25:1566-1575. [DOI: 10.1038/s41591-019-0601-5] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 08/29/2019] [Indexed: 01/12/2023]
|
28
|
Kawakami Y, Hambright WS, Takayama K, Mu X, Lu A, Cummins JH, Matsumoto T, Yurube T, Kuroda R, Kurosaka M, Fu FH, Robbins PD, Niedernhofer LJ, Huard J. Rapamycin Rescues Age-Related Changes in Muscle-Derived Stem/Progenitor Cells from Progeroid Mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:64-76. [PMID: 31312666 PMCID: PMC6610712 DOI: 10.1016/j.omtm.2019.05.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022]
Abstract
Aging-related loss of adult stem cell function contributes to impaired tissue regeneration. Mice deficient in zinc metalloproteinase STE24 (Zmpste24−/−) exhibit premature age-related musculoskeletal pathologies similar to those observed in children with Hutchinson-Gilford progeria syndrome (HGPS). We have reported that muscle-derived stem/progenitor cells (MDSPCs) isolated from Zmpste24−/− mice are defective in their proliferation and differentiation capabilities in culture and during tissue regeneration. The mechanistic target of rapamycin complex 1 (mTORC1) regulates cell growth, and inhibition of the mTORC1 pathway extends the lifespan of several animal species. We therefore hypothesized that inhibition of mTORC1 signaling would rescue the differentiation defects observed in progeroid MDSPCs. MDSPCs were isolated from Zmpste24−/− mice, and the effects of mTORC1 on MDSPC differentiation and function were examined. We found that mTORC1 signaling was increased in senescent Zmpste24−/− MDSPCs, along with impaired chondrogenic, osteogenic, and myogenic differentiation capacity versus wild-type MDSPCs. Interestingly, we observed that mTORC1 inhibition with rapamycin improved myogenic and chondrogenic differentiation and reduced levels of apoptosis and senescence in Zmpste24−/− MDSPCs. Our results demonstrate that age-related adult stem/progenitor cell dysfunction contributes to impaired regenerative capacities and that mTORC1 inhibition may represent a potential therapeutic strategy for improving differentiation capacities of senescent stem and muscle progenitor cells.
Collapse
Affiliation(s)
- Yohei Kawakami
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - William S Hambright
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Koji Takayama
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Xiaodong Mu
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Aiping Lu
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - James H Cummins
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Takashi Yurube
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Masahiro Kurosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Freddie H Fu
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Paul D Robbins
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN 55455, USA
| | - Laura J Niedernhofer
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN 55455, USA
| | - Johnny Huard
- Steadman Philippon Research Institute, Vail, CO 81657, USA
| |
Collapse
|
29
|
Zhang M, Liu F, Zhou P, Wang Q, Xu C, Li Y, Bian L, Liu Y, Zhou J, Wang F, Yao Y, Fang Y, Li D. The MTOR signaling pathway regulates macrophage differentiation from mouse myeloid progenitors by inhibiting autophagy. Autophagy 2019; 15:1150-1162. [PMID: 30724690 DOI: 10.1080/15548627.2019.1578040] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Understanding of the mechanism for myeloid differentiation provides important insights into the hematopoietic developmental processes. By using an ESC-derived myeloid progenitor cell model, we found that CSF2/GM-CSF triggered macrophage differentiation and activation of the MTOR signaling pathway. Activation or inhibition of the MTOR signaling enhanced or attenuated macrophage differentiation, respectively, suggesting a critical function. We further showed that macroautophagy/autophagy was inhibited with the addition of CSF2. Furthermore, pharmacological inhibition and genetic modification of autophagy enhanced macrophage differentiation and rescued the inhibitory effect on differentiation caused by MTOR inhibition. Thus, the MTOR signaling pathway regulates macrophage differentiation of myeloid progenitors by inhibiting autophagy. Our results provide new insights into the mechanisms for myeloid differentiation and may prove useful for therapeutic applications of hematopoietic and myeloid progenitor cells. Abbreviations: 2-DG: 2-deoxy-D-glucose; ADGRE1/F4/80: adhesion G protein-coupled receptor E1; BM: bone marrow; CQ: chloroquine; ECAR: extracellular acidification rate; ESC: embryonic stem cell; CSF2/GM-CSF: colony stimulating factor 2; CSF3/G-CSF: colony stimulating factor 3; HPC: hematopoietic progenitor cell; ITGAM/CD11b: integrin alpha M; LPS: lipopolysaccharide; MFI: median fluorescence intensity; MTOR: mechanistic target of rapamycin kinase; RPS6KB1/p70S6K1: ribosomal protein S6 kinase, polypeptide 1; shRNA: short hairpin RNA; SQSTM1/p62: sequestosome 1.
Collapse
Affiliation(s)
- Meichao Zhang
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Furao Liu
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Pingting Zhou
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Qian Wang
- b Department of Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Ci Xu
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Yanyan Li
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Lei Bian
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Yuanhua Liu
- c Department of Chemotherapy , Nanjing Medical University Affiliated Cancer Hospital, Cancer Institute of Jiangsu Province , Nanjing , Jiangsu , China
| | - Jiaxi Zhou
- d State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin , China
| | - Fei Wang
- e Department of Cell and Developmental Biology , University of Illinois at Urbana-Champaign , Urbana , IL , USA
| | - Yuan Yao
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Yong Fang
- f Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital , Shanghai JiaoTong University School of Medicine , Shanghai , China
| | - Dong Li
- a Department of Radiation Oncology, Shanghai Ninth People's Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| |
Collapse
|
30
|
Tan P, Wang H, Zhan J, Ma X, Cui X, Wang Y, Wang Y, Zhong J, Liu Y. Rapamycin‑induced miR‑30a downregulation inhibits senescence of VSMCs by targeting Beclin1. Int J Mol Med 2019; 43:1311-1320. [PMID: 30747228 PMCID: PMC6365076 DOI: 10.3892/ijmm.2019.4074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 01/16/2019] [Indexed: 12/27/2022] Open
Abstract
Vascular senescence is considered to be an independent risk factor for cardiovascular diseases. The present study aimed to investigate the effects of rapamycin on miR‑30a and its relationship with autophagy and senescence in vascular smooth muscle cells (VSMCs). Young and aging VSMCs were treated with rapamycin or transfected with miR‑30a mimics. Measurement of cellular senescence was conducted using senescence‑associated (SA)‑β‑Galactosidase (gal) staining. Dual luciferase reporter assay was used to confirm binding for miR‑30a and Beclin1. The expression levels of miR‑30a and Beclin1 were determined with reverse transcription‑quantitative polymerase chain reaction analysis. Autophagy‑related protein levels were determined using immunofluorescence or western blot assays. The results demonstrated that rapamycin treatment significantly decreased miR‑30a expression and increased Beclin1 expression in both young and aging cells, as well as promoted autophagy in VSMCs. In addition, rapamycin inhibited senescence in VSMCs and could also alleviate the aging VSMC cycle arrest. Dual luciferase reporter assay confirmed that miR‑30a could directly bind the 3'untranslated region of Beclin1 and inhibit its expression. Furthermore, miR‑30a inhibited autophagy and promoted senescence of VSMCs. In conclusion, the present results indicated that rapamycin could inhibit the senescence of VSMCs by downregulating miR‑30a, which resulted in upregulation of Beclin1 and activation of autophagy. The current study is the first to demonstrate an inhibitory role of rapamycin on VSMC senescence and might provide novel insights and potential new molecular targets in senescence treatment.
Collapse
Affiliation(s)
- Pan Tan
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Haiqin Wang
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Junkun Zhan
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Xinyu Ma
- Department of Anesthesiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Xingjun Cui
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yanjiao Wang
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yi Wang
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Jiayu Zhong
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Youshuo Liu
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
31
|
Abstract
Rapamycin inhibits cell proliferation, yet preserves (re)-proliferative potential (RPP). RPP is a potential of quiescent cells that is lost in senescent cells. mTOR drives conversion from quiescence to senescence (geroconversion). By suppressing geroconversion, rapamycin preserves RPP. Geroconversion is characterized by proliferation-like levels of phospho-S6K/S6/4E-BP1 in nonproliferating cells arrested by p16 and/or p21. mTOR-driven geroconversion is associated with cellular hyperfunction, which in turn leads to organismal aging manifested by age-related diseases.
Collapse
|
32
|
Malik N, Sansom OJ, Michie AM. The role of mTOR-mediated signals during haemopoiesis and lineage commitment. Biochem Soc Trans 2018; 46:1313-1324. [PMID: 30154096 PMCID: PMC6195642 DOI: 10.1042/bst20180141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
The serine/threonine protein kinase mechanistic target of rapamycin (mTOR) has been implicated in the regulation of an array of cellular functions including protein and lipid synthesis, proliferation, cell size and survival. Here, we describe the role of mTOR during haemopoiesis within the context of mTORC1 and mTORC2, the distinct complexes in which it functions. The use of conditional transgenic mouse models specifically targeting individual mTOR signalling components, together with selective inhibitors, have generated a significant body of research emphasising the critical roles played by mTOR, and individual mTOR complexes, in haemopoietic lineage commitment and development. This review will describe the profound role of mTOR in embryogenesis and haemopoiesis, underscoring the importance of mTORC1 at the early stages of haemopoietic cell development, through modulation of stem cell potentiation and self-renewal, and erythroid and B cell lineage commitment. Furthermore, the relatively discrete role of mTORC2 in haemopoiesis will be explored during T cell development and B cell maturation. Collectively, this review aims to highlight the functional diversity of mTOR signalling and underline the importance of this pathway in haemopoiesis.
Collapse
Affiliation(s)
- Natasha Malik
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, U.K
| | - Owen J Sansom
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, U.K
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, U.K
| | - Alison M Michie
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, U.K.
| |
Collapse
|
33
|
Ianniciello A, Rattigan KM, Helgason GV. The Ins and Outs of Autophagy and Metabolism in Hematopoietic and Leukemic Stem Cells: Food for Thought. Front Cell Dev Biol 2018; 6:120. [PMID: 30320108 PMCID: PMC6169402 DOI: 10.3389/fcell.2018.00120] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/05/2018] [Indexed: 12/11/2022] Open
Abstract
Discovered over fifty years ago, autophagy is a double-edged blade. On one hand, it regulates cellular energy sources by "cannibalization" of its own cellular components, feeding on proteins and other unused cytoplasmic factors. On the other, it is a recycling process that removes dangerous waste from the cytoplasm keeping the cell clean and healthy. Failure of the autophagic machinery is translated in dysfunction of the immune response, in aging, and in the progression of pathologies such as Parkinson disease, diabetes, and cancer. Further investigation identified autophagy with a protective role in specific types of cancer, whereas in other cases it can promote tumorigenesis. Evidence shows that treatment with chemotherapeutics can upregulate autophagy in order to maintain a stable intracellular environment promoting drug resistance and cell survival. Leukemia, a blood derived cancer, represents one of the malignancies in which autophagy is responsible for drug treatment failure. Inhibition of autophagy is becoming a strategic target for leukemic stem cell (LSC) eradication. Interestingly, the latest findings demonstrate that LSCs show higher levels of mitochondrial metabolism compared to normal stem cells. With this review, we aim to explore the links between autophagy and metabolism in the hematopoietic system, with special focus on primitive LSCs.
Collapse
Affiliation(s)
| | | | - G. Vignir Helgason
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
34
|
Abstract
Inhibitors of mTOR, including clinically available rapalogs such as rapamycin (Sirolimus) and Everolimus, are gerosuppressants, which suppress cellular senescence. Rapamycin slows aging and extends life span in a variety of species from worm to mammals. Rapalogs can prevent age-related diseases, including cancer, atherosclerosis, obesity, neurodegeneration and retinopathy and potentially rejuvenate stem cells, immunity and metabolism. Here, I further suggest how rapamycin can be combined with metformin, inhibitors of angiotensin II signaling (Losartan, Lisinopril), statins (simvastatin, atorvastatin), propranolol, aspirin and a PDE5 inhibitor. Rational combinations of these drugs with physical exercise and an anti-aging diet (Koschei formula) can maximize their anti-aging effects and decrease side effects.
Collapse
|
35
|
Abstract
The mammalian target of rapamycin (mTOR) senses nutrients and growth factors to coordinate cell growth, metabolism and autophagy. Extensive research has mapped the signaling pathways regulated by mTOR that are involved in human diseases, such as cancer, and in diabetes and ageing. Recently, however, new studies have demonstrated important roles for mTOR in promoting the differentiation of adult stem cells, driving the growth and proliferation of stem and progenitor cells, and dictating the differentiation program of multipotent stem cell populations. Here, we review these advances, providing an overview of mTOR signaling and its role in murine and human stem and progenitor cells.
Collapse
Affiliation(s)
- Delong Meng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anderson R Frank
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jenna L Jewell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA .,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
36
|
Selman C, Sinclair A, Pedroni SMA, Irvine EE, Michie AM, Withers DJ. Evidence that hematopoietic stem cell function is preserved during aging in long-lived S6K1 mutant mice. Oncotarget 2017; 7:29937-43. [PMID: 27083004 PMCID: PMC5058654 DOI: 10.18632/oncotarget.8729] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 04/05/2016] [Indexed: 11/25/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) signalling pathway plays a highly conserved role in aging; mice lacking ribosomal protein S6 kinase 1 (S6K1−/−) have extended lifespan and healthspan relative to wild type (WT) controls. Exactly how reduced mTOR signalling induces such effects is unclear, although preservation of stem cell function may be important. We show, using gene expression analyses, that there was a reduction in expression of cell cycle genes in young (12 week) and aged (80 week) S6K1−/− BM-derived c-Kit+ cells when compared to age-matched WT mice, suggesting that these cells are more quiescent in S6K1−/− mice. In addition, we investigated hematopoietic stem cell (HSC) frequency and function in young and aged S6K1−/− and WT mice. Young, but not aged, S6K1−/− mice had more LSK (lineage−, c-Kit+, Sca-1+) cells (% of bone marrow (BM)), including the most primitive long-term repopulating HSCs (LT-HSC) relative to WT controls. Donor-derived engraftment of LT-HSCs in recipient mice was unaffected by genotype in young mice, but was enhanced in transplants using LT-HSCs derived from aged S6K1−/− mice. Our results are the first to provide evidence that age-associated HSC functional decline is ameliorated in a long-lived mTOR mutant mouse.
Collapse
Affiliation(s)
- Colin Selman
- Glasgow Ageing Research Network (GARNER), Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Amy Sinclair
- Glasgow Ageing Research Network (GARNER), Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Silvia M A Pedroni
- Metabolic Signaling Group, Medical Research Council Clinical Sciences Centre, Imperial College, London, UK
| | - Elaine E Irvine
- Metabolic Signaling Group, Medical Research Council Clinical Sciences Centre, Imperial College, London, UK
| | - Alison M Michie
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Dominic J Withers
- Metabolic Signaling Group, Medical Research Council Clinical Sciences Centre, Imperial College, London, UK
| |
Collapse
|
37
|
Proto JD, Lu A, Dorronsoro A, Scibetta A, Robbins PD, Niedernhofer LJ, Huard J. Inhibition of NF-κB improves the stress resistance and myogenic differentiation of MDSPCs isolated from naturally aged mice. PLoS One 2017. [PMID: 28640861 PMCID: PMC5480862 DOI: 10.1371/journal.pone.0179270] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
A decline in the regenerative capacity of adult stem cells with aging is well documented. As a result of this decline, the efficacy of autologous stem cell therapies is likely to decline with increasing donor age. In these cases, strategies to restore the function of aged stem cells would have clinical utility. Globally, the transcription factor NF-κB is up-regulated in aged tissues. Given the negative role that NF-κB plays in myogenesis, we investigated whether the age-related decline in the function of muscle-derived stem/progenitor cells (MDSPCs) could be improved by inhibition of NF-κB. Herein, we demonstrate that pharmacologic or genetic inhibition of NF-κB activation increases myogenic differentiation and improves resistance to oxidative stress. Our results suggest that MDSPC “aging” may be reversible, and that pharmacologic targeting of pathways such as NF-κB may enhance the efficacy of cell-based therapies.
Collapse
Affiliation(s)
- Jonathan D. Proto
- Department of Medicine, Division of Molecular Medicine, Columbia University, New York, NY, United States of America
| | - Aiping Lu
- Department of Orthopaedic Surgery, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, United States of America
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO, United States of America
| | - Akaitz Dorronsoro
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL, United States of America
| | - Alex Scibetta
- Department of Orthopaedic Surgery, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, United States of America
| | - Paul D. Robbins
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL, United States of America
| | - Laura J. Niedernhofer
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL, United States of America
| | - Johnny Huard
- Department of Orthopaedic Surgery, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, United States of America
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO, United States of America
- * E-mail:
| |
Collapse
|
38
|
Leontieva OV, Blagosklonny MV. While reinforcing cell cycle arrest, rapamycin and Torins suppress senescence in UVA-irradiated fibroblasts. Oncotarget 2017; 8:109848-109856. [PMID: 29312653 PMCID: PMC5752566 DOI: 10.18632/oncotarget.17827] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 05/02/2017] [Indexed: 12/20/2022] Open
Abstract
Sunlight predisposes to skin cancer and melanomas. Ultraviolet A (UVA), a long wave component of sunlight, can reach dermal fibroblasts. Here we studied UVA-induced senescence in human fibroblasts in vitro. It is known that senescence occurs, when cell cycle is arrested, but mTOR is still active, thus converting arrest to senescence (geroconversion). We showed that, while arresting cell cycle, UVA did not inhibit mTOR, enabling geroconversion. In UVA-treated cells, mTOR remained fully active. Rapamycin and Torins 1/ 2 prevented UVA-induced senescent phenotype, although they further re-enforced cell cycle arrest. Given that senescent stromal fibroblasts support tumorigenesis, we envision that mTOR inhibitors may potentially be used to prevent sunlight-caused tumors as well as skin photo-aging.
Collapse
Affiliation(s)
- Olga V Leontieva
- Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | |
Collapse
|
39
|
Ghosh J, Kapur R. Role of mTORC1-S6K1 signaling pathway in regulation of hematopoietic stem cell and acute myeloid leukemia. Exp Hematol 2017; 50:13-21. [PMID: 28342808 DOI: 10.1016/j.exphem.2017.02.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/13/2017] [Accepted: 02/24/2017] [Indexed: 01/07/2023]
Abstract
Dysregulation of the mechanistic target of rapamycin complex 1 (mTORC1)-p70 ribosomal protein kinase 1 (S6K1) signaling pathway occurs frequently in acute myeloid leukemia (AML) patients. This pathway also plays a critical role in maintaining normal cellular processes. Given the importance of leukemia stem cells (LSCs) in the development of minimal residual disease, it is critical to use therapeutic interventions that target the LSC population to prevent disease relapse. The mTORC1-S6K1 pathway has been identified as an important regulator of hematopoietic stem cell (HSC) and LSC functions. Both HSC and LSC functions require regulation of key cellular processes including proliferation, metabolism, and autophagy, which are regulated by mTORC1 pathway. Despite the mTORC1-S6K1 pathway being a critical regulator of AML initiation and progression, inhibitors of this pathway alone have yielded mixed results in clinical studies. Recent studies have identified strategies to develop new mTORC1-S6K1 inhibitors such as RapaLink-1, which could circumvent the drug resistance observed in AML cells and in LSCs. Here, we review recent advances made in identifying the role of different components of this pathway in the regulation of HSCs and LSCs and discuss possible therapeutic approaches.
Collapse
Affiliation(s)
- Joydeep Ghosh
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Reuben Kapur
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
40
|
Yucel D, Kocabas F. Developments in Hematopoietic Stem Cell Expansion and Gene Editing Technologies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1079:103-125. [DOI: 10.1007/5584_2017_114] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
41
|
Ghosh J, Kapur R. Regulation of Hematopoietic Stem Cell Self-Renewal and Leukemia Maintenance by the PI3K-mTORC1 Pathway. CURRENT STEM CELL REPORTS 2016. [DOI: 10.1007/s40778-016-0067-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
42
|
Gabrion A, Hmitou I, Moshous D, Neven B, Lefèvre-Utile A, Diana JS, Suarez F, Picard C, Blanche S, Fischer A, Cavazzana M, Touzot F. Mammalian target of rapamycin inhibition counterbalances the inflammatory status of immune cells in patients with chronic granulomatous disease. J Allergy Clin Immunol 2016; 139:1641-1649.e6. [PMID: 27702670 DOI: 10.1016/j.jaci.2016.08.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/13/2016] [Accepted: 08/12/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Chronic granulomatous disease (CGD) is a primary immunodeficiency caused by defective production of reactive oxygen species in phagocytic cells that results in life-threatening infections and severe inflammatory manifestations. The treatment of inflammatory manifestations remains challenging because it can be associated with an increased risk of infections. Previous studies have shown that phagocytes from patients with CGD display a defect in autophagy and a reactive oxygen species-independent activation of the inflammasome. OBJECTIVE Because the intersections between autophagy and the inflammasome have been observed in patients with various diseases and microbial infections, we investigated the possible benefit of restoring the autophagy defect through rapamycin, a potent autophagy inducer, in the setting of CGD. METHODS We studied 15 patients given a diagnosis of CGD and followed in our institution. All patients were free of any active infection at the time of the study. RESULTS We show that patients with CGD present a consistent inflammatory phenotype defined by (1) increased nonclassical and intermediate monocytes, (2) a proinflammatory state of mononuclear phagocytes with increased IL-1β and TNF-α content, (3) a TH17 bias of CD4+ T cells, (4) and an increase in IL-17A-secreting neutrophil numbers. We document the reversion of CGD inflammatory status by the mammalian target of rapamycin inhibitor rapamycin on the different immune cell subsets. We also provide evidence for the enhancement of rapamycin's inhibitory effect on IL-1β secretion by the IL-1 receptor antagonist anakinra in phagocytes of patients with CGD. CONCLUSION Altogether, these data open new therapeutic approaches for CGD-related inflammatory manifestations.
Collapse
Affiliation(s)
- Aurélie Gabrion
- Biotherapy Department, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris, AP-HP, Paris, France
| | - Isabelle Hmitou
- Biotherapy Department, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris, AP-HP, Paris, France
| | - Despina Moshous
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, AP-HP, Paris, France; University Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France; Inserm UMR 1163, Paris, France
| | - Bénédicte Neven
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, AP-HP, Paris, France; University Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France; Inserm UMR 1163, Paris, France
| | - Alain Lefèvre-Utile
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Jean-Sébastien Diana
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Félipe Suarez
- University Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France; Inserm UMR 1163, Paris, France; Department of Hematology, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Capucine Picard
- University Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France; Inserm UMR 1163, Paris, France; Centre d'Etude des Déficits Immunitaires, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Stéphane Blanche
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, AP-HP, Paris, France; University Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Alain Fischer
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, AP-HP, Paris, France; University Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France; Inserm UMR 1163, Paris, France; Collège de France, Paris, France
| | - Marina Cavazzana
- Biotherapy Department, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris, AP-HP, Paris, France; University Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France; Inserm UMR 1163, Paris, France
| | - Fabien Touzot
- Biotherapy Department, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris, AP-HP, Paris, France; University Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France; Inserm UMR 1163, Paris, France.
| |
Collapse
|
43
|
Leontieva OV, Demidenko ZN, Blagosklonny MV. Dual mTORC1/C2 inhibitors suppress cellular geroconversion (a senescence program). Oncotarget 2016; 6:23238-48. [PMID: 26177051 PMCID: PMC4695114 DOI: 10.18632/oncotarget.4836] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 07/24/2015] [Indexed: 11/30/2022] Open
Abstract
In proliferating cells, mTOR is active and promotes cell growth. When the cell cycle is arrested, then mTOR converts reversible arrest to senescence (geroconversion). Rapamycin and other rapalogs suppress geroconversion, maintaining quiescence instead. Here we showed that ATP-competitive kinase inhibitors (Torin1 and PP242), which inhibit both mTORC1 and TORC2, also suppressed geroconversion. Despite inhibition of proliferation (in proliferating cells), mTOR inhibitors preserved re-proliferative potential (RP) in arrested cells. In p21-arrested cells, Torin 1 and PP242 detectably suppressed geroconversion at concentrations as low as 1-3 nM and 10-30 nM, reaching maximal gerosuppression at 30 nM and 300 nM, respectively. Near-maximal gerosuppression coincided with inhibition of p-S6K(T389) and p-S6(S235/236). Dual mTOR inhibitors prevented senescent morphology and hypertrophy. Our study warrants investigation into whether low doses of dual mTOR inhibitors will prolong animal life span and delay age-related diseases. A new class of potential anti-aging drugs can be envisioned.
Collapse
Affiliation(s)
- Olga V Leontieva
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Zoya N Demidenko
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | |
Collapse
|
44
|
Chung EJ, Sowers A, Thetford A, McKay-Corkum G, Chung SI, Mitchell JB, Citrin DE. Mammalian Target of Rapamycin Inhibition With Rapamycin Mitigates Radiation-Induced Pulmonary Fibrosis in a Murine Model. Int J Radiat Oncol Biol Phys 2016; 96:857-866. [PMID: 27663762 DOI: 10.1016/j.ijrobp.2016.07.026] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/18/2016] [Accepted: 07/21/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE Radiation-induced pulmonary fibrosis (RIPF) is a late toxicity of therapeutic radiation. Signaling of the mammalian target of rapamycin drives several processes implicated in RIPF, including inflammatory cytokine production, fibroblast proliferation, and epithelial senescence. We sought to determine if mammalian target of rapamycin inhibition with rapamycin would mitigate RIPF. METHODS AND MATERIALS C57BL/6NCr mice received a diet formulated with rapamycin (14 mg/kg food) or a control diet 2 days before and continuing for 16 weeks after exposure to 5 daily fractions of 6 Gy of thoracic irradiation. Fibrosis was assessed with Masson trichrome staining and hydroxyproline assay. Cytokine expression was evaluated by quantitative real-time polymerase chain reaction. Senescence was assessed by staining for β-galactosidase activity. RESULTS Administration of rapamycin extended the median survival of irradiated mice compared with the control diet from 116 days to 156 days (P=.006, log-rank test). Treatment with rapamycin reduced hydroxyproline content compared with the control diet (irradiation plus vehicle, 45.9 ± 11.8 μg per lung; irradiation plus rapamycin, 21.4 ± 6.0 μg per lung; P=.001) and reduced visible fibrotic foci. Rapamycin treatment attenuated interleukin 1β and transforming growth factor β induction in irradiated lungs compared with the control diet. Type II pneumocyte senescence after irradiation was reduced with rapamycin treatment at 16 weeks (3-fold reduction at 16 weeks, P<.001). CONCLUSIONS Rapamycin protected against RIPF in a murine model. Rapamycin treatment reduced inflammatory cytokine expression, extracellular matrix production, and senescence in type II pneumocytes.
Collapse
Affiliation(s)
- Eun Joo Chung
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Anastasia Sowers
- Radiation Biology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Angela Thetford
- Radiation Biology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Grace McKay-Corkum
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Su I Chung
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Deborah E Citrin
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
45
|
Rejuvenating immunity: "anti-aging drug today" eight years later. Oncotarget 2016; 6:19405-12. [PMID: 25844603 PMCID: PMC4637294 DOI: 10.18632/oncotarget.3740] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/28/2015] [Indexed: 01/02/2023] Open
Abstract
The 2014 year ended with celebration: Everolimus, a rapamycin analog, was shown to improve immunity in old humans, heralding ‘a turning point’ in research and new era in human quest for immortality. Yet, this turning point was predicted a decade ago. But what will cause human death, when aging will be abolished?
Collapse
|
46
|
Alessio N, Del Gaudio S, Capasso S, Di Bernardo G, Cappabianca S, Cipollaro M, Peluso G, Galderisi U. Low dose radiation induced senescence of human mesenchymal stromal cells and impaired the autophagy process. Oncotarget 2016; 6:8155-66. [PMID: 25544750 PMCID: PMC4480742 DOI: 10.18632/oncotarget.2692] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 11/02/2014] [Indexed: 12/20/2022] Open
Abstract
Low doses of radiation may have profound effects on cellular function. Individuals may be exposed to low doses of radiation either intentionally for medical purposes or accidentally, such as those exposed to radiological terrorism or those who live near illegal radioactive waste dumpsites.We studied the effects of low dose radiation on human bone marrow mesenchymal stromal cells (MSC), which contain a subpopulation of stem cells able to differentiate in bone, cartilage, and fat; support hematopoiesis; and contribute to body's homeostasis.The main outcome of low radiation exposure, besides reduction of cell cycling, is the triggering of senescence, while the contribution to apoptosis is minimal. We also showed that low radiation affected the autophagic flux. We hypothesize that the autophagy prevented radiation deteriorative processes, and its decline contributed to senescence.An increase in ATM staining one and six hours post-irradiation and return to basal level at 48 hours, along with persistent gamma-H2AX staining, indicated that MSC properly activated the DNA repair signaling, though some damages remained unrepaired, mainly in non-cycling cells. This suggested that the impaired DNA repair capacity of irradiated MSC seemed mainly related to the reduced activity of a non-homologous end-joining (NHEJ) system rather than HR (homologous recombination).
Collapse
Affiliation(s)
- Nicola Alessio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples 80138, Italy
| | - Stefania Del Gaudio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples 80138, Italy
| | - Stefania Capasso
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples 80138, Italy
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples 80138, Italy
| | - Salvatore Cappabianca
- Department "F. Magrassi - A. Lanzara" Second University of Naples, Naples 80138, Italy
| | - Marilena Cipollaro
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples 80138, Italy
| | | | - Umberto Galderisi
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA 19107, USA.,Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples 80138, Italy.,Institute of Bioscience and Bioresources, CNR, Naples 80138, Italy
| |
Collapse
|
47
|
Fafián-Labora J, Fernández-Pernas P, Fuentes I, De Toro J, Oreiro N, Sangiao-Alvarellos S, Mateos J, Arufe M. Influence of age on rat bone-marrow mesenchymal stem cells potential. Sci Rep 2015; 5:16765. [PMID: 26581954 PMCID: PMC4652164 DOI: 10.1038/srep16765] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 10/20/2015] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stem cells promising role in cell-based therapies and tissue engineering appears to be limited due to a decline of their regenerative potential with increasing donor age. Six age groups from bone marrow mesenchymal stem cells of Wistar rats were studied (newborn, infant, young, pre-pubertal, pubertal and adult). Quantitative proteomic assay was performance by iTRAQ using an 8-plex iTRAQ labeling and the proteins differentially expressed were grouped in pluripotency, proliferative and metabolism processes. Proliferation makers, CD117 and Ki67 were measure by flow cytometry assay. Real time polymerase chain reaction analysis of pluripotency markers Rex1, Oct4, Sox2 and Nanog were done. Biological differentiation was realized using specific mediums for 14 days to induce osteogenesis, adipogenesis or chondrogenesis and immunostain analysis of differentiated cell resulting were done. Enzimoimmunoassay analysis of several enzymes as L-lactate dehydrogenase and glucose-6-phosphate isomerase were also done to validate iTRAQ data. Taking together these results indicate for the first time that mesenchymal stem cells have significant differences in their proliferative, pluripotency and metabolism profiles and those differences are age depending.
Collapse
Affiliation(s)
- J. Fafián-Labora
- Grupo de Terapia Celular y Medicina Regenerativa (TCMR-CHUAC). CIBER-BBN/ISCIII. Servicio de Reumatología. Instituto de Investigación Biomédica de A Coruña (INIBIC). Complexo Hospitalario Universitario de A Coruña (CHUAC). SERGAS. Departamento de Medicina. Facultade de Oza. Universidade de A Coruña (UDC). As Xubias, 15006. A Coruña, Spain
| | - P. Fernández-Pernas
- Grupo de Terapia Celular y Medicina Regenerativa (TCMR-CHUAC). CIBER-BBN/ISCIII. Servicio de Reumatología. Instituto de Investigación Biomédica de A Coruña (INIBIC). Complexo Hospitalario Universitario de A Coruña (CHUAC). SERGAS. Departamento de Medicina. Facultade de Oza. Universidade de A Coruña (UDC). As Xubias, 15006. A Coruña, Spain
| | - I. Fuentes
- Grupo de Terapia Celular y Medicina Regenerativa (TCMR-CHUAC). CIBER-BBN/ISCIII. Servicio de Reumatología. Instituto de Investigación Biomédica de A Coruña (INIBIC). Complexo Hospitalario Universitario de A Coruña (CHUAC). SERGAS. Departamento de Medicina. Facultade de Oza. Universidade de A Coruña (UDC). As Xubias, 15006. A Coruña, Spain
| | - J. De Toro
- Grupo de Terapia Celular y Medicina Regenerativa (TCMR-CHUAC). CIBER-BBN/ISCIII. Servicio de Reumatología. Instituto de Investigación Biomédica de A Coruña (INIBIC). Complexo Hospitalario Universitario de A Coruña (CHUAC). SERGAS. Departamento de Medicina. Facultade de Oza. Universidade de A Coruña (UDC). As Xubias, 15006. A Coruña, Spain
| | - N. Oreiro
- Grupo de Proteómica-PBR2-ProteoRed/ISCIII-Servicio de Reumatologia. Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas. Universidade da Coruña (UDC). As Xubias, 15006. A Coruña, España
| | - S. Sangiao-Alvarellos
- Grupo Fisiopatología Endocrina, Nutricional y Médica (FENM-CHUAC). Instituto de Investigación Biomédica de A Coruña (INIBIC). Complexo Hospitalario Universitario de A Coruña (CHUAC). SERGAS. Departamento de Medicina. Facultade de Oza. Universidade de A Coruña (UDC). As Xubias, 15006. A Coruña, Spain
| | - J. Mateos
- Grupo de Terapia Celular y Medicina Regenerativa (TCMR-CHUAC). CIBER-BBN/ISCIII. Servicio de Reumatología. Instituto de Investigación Biomédica de A Coruña (INIBIC). Complexo Hospitalario Universitario de A Coruña (CHUAC). SERGAS. Departamento de Medicina. Facultade de Oza. Universidade de A Coruña (UDC). As Xubias, 15006. A Coruña, Spain
| | - M.C. Arufe
- Grupo de Terapia Celular y Medicina Regenerativa (TCMR-CHUAC). CIBER-BBN/ISCIII. Servicio de Reumatología. Instituto de Investigación Biomédica de A Coruña (INIBIC). Complexo Hospitalario Universitario de A Coruña (CHUAC). SERGAS. Departamento de Medicina. Facultade de Oza. Universidade de A Coruña (UDC). As Xubias, 15006. A Coruña, Spain
| |
Collapse
|
48
|
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: 3.4] [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
|
49
|
Abstract
Cellular senescence happens in 2 steps: cell cycle arrest followed, or sometimes preceded, by gerogenic conversion (geroconversion). Geroconvesrion is a form of growth, a futile growth during cell cycle arrest. It converts reversible arrest to irreversible senescence. Geroconversion is driven by growth-promoting, mitogen-/nutrient-sensing pathways such as mTOR. Geroconversion leads to hyper-secretory, hypertrophic and pro-inflammatory cellular phenotypes, hyperfunctions and malfunctions. On organismal level, geroconversion leads to age-related diseases and death. Rapamycin, a gerosuppressant, extends life span in diverse species from yeast to mammals. Stress-and oncogene-induced accelerated senescence, replicative senescence in vitro and life-long cellular aging in vivo all can be described by 2-step model.
Collapse
|
50
|
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
|