201
|
Migliaccio AR. A vicious interplay between genetic and environmental insults in the etiology of blood cancers. Exp Hematol 2017; 59:9-13. [PMID: 29248611 DOI: 10.1016/j.exphem.2017.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/06/2017] [Accepted: 12/07/2017] [Indexed: 02/06/2023]
Abstract
Over the years, the etiology of cancer has been attributed alternatively to genetic and environmental insults. According to the genetic hypothesis, cancer cells arise from the acquisition of mutations that dysregulate the intrinsic mechanisms controlling normal cell growth and survival. In contrast, the environmental hypothesis holds that cancer can be caused by multiple extrinsic challenges that alter normal tissue homeostasis, but may not necessarily involve mutations. It is, however, quite possible that these two mechanisms are not mutually exclusive. According to this third hypothesis, environmental challenges, by mechanisms still poorly understood, may act by imposing a selection pressure that confers a proliferative advantage on cells carrying specific driver mutations, leading to disease initiation. The authors of a brief report published in this journal (Exp Hematol. 2017;56:1-6) tested this third hypothesis experimentally and provide new evidence that chronic inflammation, by increasing tumor necrosis factor (TNF)-α, may positively select malignant hematopoietic stem cells (HSCs) carrying loss-of-function TET2 mutations that switch the TNF-α signaling responses to activate proliferation rather than inducing quiescence. Furthermore, these mutations skew hematopoietic differentiation toward the myelomonocytic lineage and the output of macrophages producing TNF-α constitutively, promoting further environment-independent expansion of the malignant HSCs. These findings support a model in which the formation of a malignant population is triggered by a vicious interplay between genetic (TET2 mutations) and environmental (inflammation) insults, suggesting the need for strategies that target both the malignant cells and their environment.
Collapse
Affiliation(s)
- Anna Rita Migliaccio
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, Bologna, Italy; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, New York.
| |
Collapse
|
202
|
The paths of mortality: how understanding the biology of aging can help explain systems behavior of single cells. ACTA ACUST UNITED AC 2017; 8:25-31. [PMID: 29552673 DOI: 10.1016/j.coisb.2017.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Aging is a fundamental aspect of life, yet also one of the most confounding. In individual cells, aging results in a progressive decline which affects all organelles and reduces a cell's ability to maintain homeostasis. Because of the interconnected nature of cellular systems, the failure of even a single organelle can have cascading effects. We are just beginning to understand the dramatic physiological changes that occur during aging. Because most aging research has focused on population dynamics, or differences between wild-type and mutant populations, single-cell behavior has been largely overlooked. An open question is whether aging cells are defined by predictable sequences of physiological changes, or whether they proceed along divergent aging trajectories defined by whichever system begins to fail first. Can aging be best characterized by a cell-cycle like model with stereotyped states all cells progress through, or a Waddington landscape with divergent trajectories? Here we present work on understanding the changing physiological states of aging cells, why it will impact systems and synthetic biologists, and how the systems community can contribute significantly to the study of aging.
Collapse
|
203
|
Nalapareddy K, Nattamai KJ, Kumar RS, Karns R, Wikenheiser-Brokamp KA, Sampson LL, Mahe MM, Sundaram N, Yacyshyn MB, Yacyshyn B, Helmrath MA, Zheng Y, Geiger H. Canonical Wnt Signaling Ameliorates Aging of Intestinal Stem Cells. Cell Rep 2017; 18:2608-2621. [PMID: 28297666 PMCID: PMC5987258 DOI: 10.1016/j.celrep.2017.02.056] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 01/10/2017] [Accepted: 02/16/2017] [Indexed: 12/30/2022] Open
Abstract
Although intestinal homeostasis is maintained by intestinal stem cells (ISCs), regeneration is impaired upon aging. Here, we first uncover changes in intestinal architecture, cell number, and cell composition upon aging. Second, we identify a decline in the regenerative capacity of ISCs upon aging because of a decline in canonical Wnt signaling in ISCs. Changes in expression of Wnts are found in stem cells themselves and in their niche, including Paneth cells and mesenchyme. Third, reactivating canonical Wnt signaling enhances the function of both murine and human ISCs and, thus, ameliorates aging-associated phenotypes of ISCs in an organoid assay. Our data demonstrate a role for impaired Wnt signaling in physiological aging of ISCs and further identify potential therapeutic avenues to improve ISC regenerative potential upon aging.
Collapse
Affiliation(s)
- Kodandaramireddy Nalapareddy
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Kalpana J Nattamai
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Rupali S Kumar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Rebekah Karns
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Kathryn A Wikenheiser-Brokamp
- Divisions of Pathology and Laboratory Medicine and Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center and Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Leesa L Sampson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Maxime M Mahe
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Nambirajan Sundaram
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Mary-Beth Yacyshyn
- Division of Digestive Diseases, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Bruce Yacyshyn
- Division of Digestive Diseases, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Michael A Helmrath
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Hartmut Geiger
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA; Institute for Molecular Medicine, Stem Cells, and Aging and Aging Research Center, Ulm University, 89081 Ulm, Germany.
| |
Collapse
|
204
|
Gruszka AM, Valli D, Alcalay M. Understanding the molecular basis of acute myeloid leukemias: where are we now? Int J Hematol Oncol 2017; 6:43-53. [PMID: 30302223 DOI: 10.2217/ijh-2017-0002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/29/2017] [Indexed: 12/26/2022] Open
Abstract
Although the treatment modalities for acute myeloid leukemia (AML) have not changed much over the past 40 years, distinct progress has been made in deciphering the basic biology underlying the pathogenesis of this group of hematological disorders. Studies show that AML development is a multicause, multistep and multipathway process. Accordingly, AMLs constitute a heterogeneous group of diseases. The thorough understanding of the molecular basis of AML is paving the way for better therapeutic approaches. Multiple novel drugs are being introduced and new, more efficient and less toxic formulations of conventional therapeutics are becoming available. Here, we review the recent advances in the comprehension of the molecular processes that lead to the onset of AML and its translation into clinical practice.
Collapse
Affiliation(s)
- Alicja M Gruszka
- Department of Experimental Oncology, Istituto Europeo di Oncologia, Via Adamello 16, 20139 Milano, Italy.,Department of Experimental Oncology, Istituto Europeo di Oncologia, Via Adamello 16, 20139 Milano, Italy
| | - Debora Valli
- Department of Experimental Oncology, Istituto Europeo di Oncologia, Via Adamello 16, 20139 Milano, Italy.,Department of Experimental Oncology, Istituto Europeo di Oncologia, Via Adamello 16, 20139 Milano, Italy
| | - Myriam Alcalay
- Department of Experimental Oncology, Istituto Europeo di Oncologia, Via Adamello 16, 20139 Milano, Italy.,Department of Oncology & Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy.,Department of Experimental Oncology, Istituto Europeo di Oncologia, Via Adamello 16, 20139 Milano, Italy.,Department of Oncology & Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milano, Italy
| |
Collapse
|
205
|
Xiong X, You C, Cao X, Pang L, Kong R, Sun X. Ni2P nanosheets array as a novel electrochemical catalyst electrode for non-enzymatic H2O2 sensing. Electrochim Acta 2017. [DOI: 10.1016/j.electacta.2017.09.104] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
206
|
Abstract
I started research in high school, experimenting on immunological tolerance to transplantation antigens. This led to studies of the thymus as the site of maturation of T cells, which led to the discovery, isolation, and clinical transplantation of purified hematopoietic stem cells (HSCs). The induction of immune tolerance with HSCs has led to isolation of other tissue-specific stem cells for regenerative medicine. Our studies of circulating competing germline stem cells in colonial protochordates led us to document competing HSCs. In human acute myelogenous leukemia we showed that all preleukemic mutations occur in HSCs, and determined their order; the final mutations occur in a multipotent progenitor derived from the preleukemic HSC clone. With these, we discovered that CD47 is an upregulated gene in all human cancers and is a "don't eat me" signal; blocking it with antibodies leads to cancer cell phagocytosis. CD47 is the first known gene common to all cancers and is a target for cancer immunotherapy.
Collapse
Affiliation(s)
- Irving Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, and Ludwig Center for Cancer Stem Cell Research and Medicine at Stanford, Stanford, CA 94305
| |
Collapse
|
207
|
Conese M, Carbone A, Beccia E, Angiolillo A. The Fountain of Youth: A Tale of Parabiosis, Stem Cells, and Rejuvenation. Open Med (Wars) 2017; 12:376-383. [PMID: 29104943 PMCID: PMC5662775 DOI: 10.1515/med-2017-0053] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 06/19/2017] [Indexed: 01/10/2023] Open
Abstract
Transfusion (or drinking) of blood or of its components has been thought as a rejuvenation method since ancient times. Parabiosis, the procedure of joining two animals so that they share each others blood circulation, has revitalized the concept of blood as a putative drug. Since 2005, a number of papers have reported the anti-ageing effect of heterochronic parabiosis, which is joining an aged mouse to a young partner. The hallmark of aging is the decline of regenerative properties in most tissues, partially attributed to impaired function of stem and progenitor cells. In the parabiosis experiments, it was elegantly shown that factors derived from the young systemic environment are able to activate molecular signaling pathways in hepatic, muscle or neural stem cells of the old parabiont leading to increased tissue regeneration. Eventually, further studies have brought to identify some soluble factors in part responsible for these rejuvenating effects, including the chemokine CCL11, the growth differentiation factor 11, a member of the TGF-β superfamily, and oxytocin. The question about giving whole blood or specific factors in helping rejuvenation is open, as well as the mechanisms of action of these factors, deserving further studies to be translated into the life of (old) human beings.
Collapse
Affiliation(s)
- Massimo Conese
- Biomedical Research Center "E. Altomare", Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, c/o Ospedali Riuniti, Via L. Pinto 1, 71122, Tel.: +39 0881 588014; ;Foggia, Italy
| | - Annalucia Carbone
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Elisa Beccia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.,Dipartimento di Medicina e Scienze della Salute "V. Tiberio", University of Molise, Campobasso, Italy
| | - Antonella Angiolillo
- Dipartimento di Medicina e Scienze della Salute "V. Tiberio", University of Molise, Campobasso, Italy
| |
Collapse
|
208
|
Keyes BE, Fuchs E. Stem cells: Aging and transcriptional fingerprints. J Cell Biol 2017; 217:79-92. [PMID: 29070608 PMCID: PMC5748991 DOI: 10.1083/jcb.201708099] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 12/31/2022] Open
Abstract
Keyes and Fuchs discuss the decline in stem cell renewal and function with aging and the ensuing consequences on tissue homeostasis and regeneration. Stem cells are imbued with unique qualities. They have the capacity to propagate themselves through symmetric divisions and to divide asymmetrically to engender new cells that can progress to differentiate into tissue-specific, terminal cell types. Armed with these qualities, stem cells in adult tissues are tasked with replacing decaying cells and regenerating tissue after injury to maintain optimal tissue function. With increasing age, stem cell functional abilities decline, resulting in reduced organ function and delays in tissue repair. Here, we review the effect of aging in five well-studied adult murine stem cell populations and explore age-related declines in stem cell function and their consequences for stem cell self-renewal, tissue homeostasis, and regeneration. Finally, we examine transcriptional changes that have been documented in aged stem cell populations and discuss new questions and future directions that this collection of data has uncovered.
Collapse
Affiliation(s)
| | - Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY
| |
Collapse
|
209
|
Rajan TS, Scionti D, Diomede F, Piattelli A, Bramanti P, Mazzon E, Trubiani O. Prolonged Expansion Induces Spontaneous Neural Progenitor Differentiation from Human Gingiva-Derived Mesenchymal Stem Cells. Cell Reprogram 2017; 19:389-401. [PMID: 29058474 DOI: 10.1089/cell.2017.0012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Neural crest-derived mesenchymal stem cells (MSCs) obtained from dental tissues received considerable interest in regenerative medicine, particularly in nerve regeneration owing to their embryonic origin and ease of harvest. Proliferation efficacy and differentiation capacity into diverse cell lineages propose dental MSCs as an in vitro tool for disease modeling. In this study, we investigated the spontaneous differentiation efficiency of dental MSCs obtained from human gingiva tissue (hGMSCs) into neural progenitor cells after extended passaging. At passage 41, the morphology of hGMSCs changed from typical fibroblast-like shape into sphere-shaped cells with extending processes. Next-generation transcriptomics sequencing showed increased expression of neural progenitor markers such as NES, MEIS2, and MEST. In addition, de novo expression of neural precursor genes, such as NRN1, PHOX2B, VANGL2, and NTRK3, was noticed in passage 41. Immunocytochemistry results showed suppression of neurogenesis repressors TP53 and p21, whereas Western blot results revealed the expression of neurotrophic factors BDNF and NT3 at passage 41. Our results showed the spontaneous efficacy of hGMSCs to differentiate into neural precursor cells over prolonged passages and that these cells may assist in producing novel in vitro disease models that are associated with neural development.
Collapse
Affiliation(s)
| | - Domenico Scionti
- 1 Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino-Pulejo," Messina , Italy
| | - Francesca Diomede
- 2 Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio," Chieti-Pescara, Chieti, Italy
| | - Adriano Piattelli
- 2 Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio," Chieti-Pescara, Chieti, Italy
| | - Placido Bramanti
- 1 Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino-Pulejo," Messina , Italy
| | - Emanuela Mazzon
- 1 Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino-Pulejo," Messina , Italy
| | - Oriana Trubiani
- 2 Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio," Chieti-Pescara, Chieti, Italy
| |
Collapse
|
210
|
Abstract
PURPOSE OF REVIEW Cell-cycle checkpoints are surveillance mechanisms in eukaryotic cells that monitor the condition of the cell, repair cellular damages, and allow the cell to progress through the various phases of the cell cycle when conditions become favorable. We review recent advances in hematopoietic stem cell (HSC) biology, highlighting a mitochondrial metabolic checkpoint that is essential for HSCs to return to the quiescent state. RECENT FINDINGS As quiescent HSCs enter the cell cycle, mitochondrial biogenesis is induced, which is associated with increased mitochondrial protein folding stress and mitochondrial oxidative stress. Mitochondrial unfolded protein response and mitochondrial oxidative stress response are activated to alleviate stresses and allow HSCs to exit the cell cycle and return to quiescence. Other mitochondrial maintenance mechanisms include mitophagy and asymmetric segregation of aged mitochondria. SUMMARY Because loss of HSC quiescence results in the depletion of the HSC pool and compromised tissue regeneration, deciphering the molecular mechanisms that regulate the mitochondrial metabolic checkpoint in HSCs will increase our understanding of hematopoiesis and how it becomes dysregulated under pathological conditions and during aging. More broadly, this knowledge is instrumental for understanding the maintenance of cells that convert between quiescence and proliferation to support their physiological functions.
Collapse
|
211
|
Zinger A, Cho WC, Ben-Yehuda A. Cancer and Aging - the Inflammatory Connection. Aging Dis 2017; 8:611-627. [PMID: 28966805 PMCID: PMC5614325 DOI: 10.14336/ad.2016.1230] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/30/2016] [Indexed: 12/13/2022] Open
Abstract
Aging and cancer are highly correlated biological phenomena. Various cellular processes such as DNA damage responses and cellular senescence that serve as tumor suppressing mechanisms throughout life result in degenerative changes and contribute to the aging phenotype. In turn, aging is considered a pro-tumorigenic state, and constitutes the single most important risk factor for cancer development. However, the causative relations between aging and cancer is not straight forward, as these processes carry contradictory hallmarks; While aging is characterized by tissue degeneration and organ loss of function, cancer is a state of sustained cellular proliferation and gain of new functions. Here, we review the molecular and cellular pathways that stand in the base of aging related cancer. Specifically, we deal with the inflammatory perspective that link these two processes, and suggest possible molecular targets that may be exploited to modify their courses.
Collapse
Affiliation(s)
- Adar Zinger
- 1Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - William C Cho
- 2Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Arie Ben-Yehuda
- 1Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
212
|
Aunan JR, Cho WC, Søreide K. The Biology of Aging and Cancer: A Brief Overview of Shared and Divergent Molecular Hallmarks. Aging Dis 2017; 8:628-642. [PMID: 28966806 PMCID: PMC5614326 DOI: 10.14336/ad.2017.0103] [Citation(s) in RCA: 226] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 01/03/2017] [Indexed: 12/17/2022] Open
Abstract
Aging is the inevitable time-dependent decline in physiological organ function and is a major risk factor for cancer development. Due to advances in health care, hygiene control and food availability, life expectancy is increasing and the population in most developed countries is shifting to an increasing proportion of people at a cancer susceptible age. Mechanisms of aging are also found to occur in carcinogenesis, albeit with shared or divergent end-results. It is now clear that aging and cancer development either share or diverge in several disease mechanisms. Such mechanisms include the role of genomic instability, telomere attrition, epigenetic changes, loss of proteostasis, decreased nutrient sensing and altered metabolism, but also cellular senescence and stem cell function. Cancer cells and aged cells are also fundamentally opposite, as cancer cells can be thought of as hyperactive cells with advantageous mutations, rapid cell division and increased energy consumption, while aged cells are hypoactive with accumulated disadvantageous mutations, cell division inability and a decreased ability for energy production and consumption. Nonetheless, aging and cancer are tightly interconnected and many of the same strategies and drugs may be used to target both, while in other cases antagonistic pleiotrophy come into effect and inhibition of one can be the activation of the other. Cancer can be considered an aging disease, though the shared mechanisms underpinning the two processes remain unclear. Better understanding of the shared and divergent pathways of aging and cancer is needed.
Collapse
Affiliation(s)
- Jan R Aunan
- 1Gastrointestinal Translational Research Unit, Molecular Lab, Stavanger University Hospital, Stavanger, Norway.,2Department of Gastrointestinal Surgery, Stavanger University Hospital, Stavanger, Norway
| | - William C Cho
- 3Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Kjetil Søreide
- 1Gastrointestinal Translational Research Unit, Molecular Lab, Stavanger University Hospital, Stavanger, Norway.,2Department of Gastrointestinal Surgery, Stavanger University Hospital, Stavanger, Norway.,4Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
213
|
Florian MC, Klenk J, Marka G, Soller K, Kiryakos H, Peter R, Herbolsheimer F, Rothenbacher D, Denkinger M, Geiger H. Expression and Activity of the Small RhoGTPase Cdc42 in Blood Cells of Older Adults Are Associated With Age and Cardiovascular Disease. J Gerontol A Biol Sci Med Sci 2017; 72:1196-1200. [PMID: 28498918 DOI: 10.1093/gerona/glx091] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Indexed: 12/11/2022] Open
Abstract
The small RhoGTPase Cdc42 is mechanistically linked to aging of multiple tissues and to rejuvenation of hematopoietic stem cells in mice. However, data validating Cdc42 activity and expression as biomarker for aging in humans are still missing. Here, we hypothesized that Cdc42 might serve as a novel biomarker of aging in older adults and therefore we determined Cdc42 activity and expression levels in peripheral blood cells from a cohort of 196 donors. We investigated the association of these parameters with both chronological and biological aging. We also tested in this cohort of older adults a recently published algorithm determining chronological age based on DNA methylation profiles. A positive correlation with chronological age was found for both the level of Cdc42 mRNA and the level of active Cdc42 protein (the GTP bound form). Notably, the level of Cdc42 mRNA as well as total protein showed a specific strong association to cardiovascular disease and Cdc42 mRNA levels also to a history of myocardial infarction. In summary, these data validate Cdc42 as a blood biomarker of both chronological aging as well as aging-associated diseases like cardiovascular disease and myocardial infarction.
Collapse
Affiliation(s)
| | - Jochen Klenk
- Institute of Epidemiology and Medical Biometry, Ulm University, Germany.,Department of Clinical Gerontology, Robert Bosch Hospital Stuttgart, Germany
| | - Gina Marka
- Institute of Molecular Medicine and Stem Cell Aging, Ulm University, Germany
| | - Karin Soller
- Institute of Molecular Medicine and Stem Cell Aging, Ulm University, Germany
| | - Hady Kiryakos
- Institute of Molecular Medicine and Stem Cell Aging, Ulm University, Germany
| | - Richard Peter
- Institute for History, Theory and Ethics in Medicine
| | | | | | - Michael Denkinger
- AGAPLESION Bethesda Clinic, Geriatric Medicine, Ulm University, Germany.,Geriatric Center Ulm/Alb-Donau, Germany
| | - Hartmut Geiger
- Institute of Molecular Medicine and Stem Cell Aging, Ulm University, Germany.,Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Ohio
| |
Collapse
|
214
|
Wahlestedt M, Bryder D. The slippery slope of hematopoietic stem cell aging. Exp Hematol 2017; 56:1-6. [PMID: 28943295 DOI: 10.1016/j.exphem.2017.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/12/2017] [Accepted: 09/15/2017] [Indexed: 10/18/2022]
Abstract
The late stages of life, in most species including humans, are associated with a decline in the overall maintenance and health of the organism. This applies also to the hematopoietic system, where aging is not only associated with an increased predisposition for hematological malignancies, but also identified as a strong comorbidity factor for other diseases. Research during the last two decades has proposed that alterations at the level of hematopoietic stem cells (HSCs) might be a root cause for the hematological changes observed with age. However, the recent realization that not all HSCs are alike with regard to fundamental stem cell properties such as self-renewal and lineage potential has several implications for HSC aging, including the synchrony and the stability of the aging HSC state. To approach HSC aging from a clonal perspective, we recently took advantage of technical developments in cellular barcoding and combined this with the derivation of induced pluripotent stem cells (iPSCs). This allowed us to selectively approach HSCs functionally affected by age. The finding that such iPSCs were capable of fully regenerating multilineage hematopoiesis upon morula/blastocyst complementation provides compelling evidence that many aspects of HSC aging can be reversed, which indicates that a central mechanism underlying HSC aging is a failure to uphold the epigenomes associated with younger age. Here we discuss these findings in the context of the underlying causes that might influence HSC aging and the requirements and prospects for restoration of the aging HSC epigenome.
Collapse
Affiliation(s)
- Martin Wahlestedt
- Medical Faculty, Institution for Laboratory Medicine, Division of Molecular Hematology, Lund University, Lund, Sweden
| | - David Bryder
- Medical Faculty, Institution for Laboratory Medicine, Division of Molecular Hematology, Lund University, Lund, Sweden; StemTherapy, Lund University, Lund, Sweden.
| |
Collapse
|
215
|
Lagunas AM, Wu J, Crowe DL. Telomere DNA damage signaling regulates cancer stem cell evolution, epithelial mesenchymal transition, and metastasis. Oncotarget 2017; 8:80139-80155. [PMID: 29113290 PMCID: PMC5655185 DOI: 10.18632/oncotarget.20960] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 08/25/2017] [Indexed: 12/16/2022] Open
Abstract
Chromosome ends are protected by telomeres that prevent DNA damage response and degradation. When telomeres become critically short, the DNA damage response is activated at chromosome ends which induces cellular senescence or apoptosis. Telomeres are protected by the double stranded DNA binding protein TRF2 and maintained by telomerase or a recombination based mechanism known as alternative lengthening of telomeres (ALT). Telomerase is expressed in the basal layer of the epidermis, and stem cells in epidermis have longer telomeres than proliferating populations. Stem cell expansion has been associated with epithelial-mesenchymal transition (EMT) in cancer. EMT is a critical process in cancer progression in which cells acquire spindle morphology, migrate from the primary tumor, and spread to distant anatomic sites. Our previous study demonstrated that loss of TRF2 expression observed in human squamous cell carcinomas expanded metastatic cancer stem cells during mouse skin carcinogenesis. To determine if telomerase inhibition could block the TRF2-null mediated expansion of metastatic clones, we characterized skin carcinogenesis in a conditional TRF2/Terc double null mutant mouse. Loss of TRF2 and Terc expression resulted in telomere DNA damage, severely depleted CD34 + and Lgr6+ cancer stem cells, and induced terminal differentiation of metastatic cancer cells. However a novel cancer stem cell population evolved in primary tumors exhibiting genomic instability, ALT, and EMT. Surprisingly we discovered that metastatic clones evolved prior to histopathologic onset of primary tumors. These results have important implications for understanding the evolution and treatment of metastatic cancer.
Collapse
Affiliation(s)
| | - Jianchun Wu
- University of Illinois Cancer Center, Chicago, IL, USA
| | - David L Crowe
- University of Illinois Cancer Center, Chicago, IL, USA
| |
Collapse
|
216
|
NRF2 Activation Impairs Quiescence and Bone Marrow Reconstitution Capacity of Hematopoietic Stem Cells. Mol Cell Biol 2017; 37:MCB.00086-17. [PMID: 28674188 DOI: 10.1128/mcb.00086-17] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 06/27/2017] [Indexed: 12/12/2022] Open
Abstract
Tissue stem cells are maintained in quiescence under physiological conditions but proliferate and differentiate to replenish mature cells under stressed conditions. The KEAP1-NRF2 system plays an essential role in stress response and cytoprotection against redox disturbance. To clarify the role of the KEAP1-NRF2 system in tissue stem cells, we focused on hematopoiesis in this study and used Keap1-deficient mice to examine the effects of persistent activation of NRF2 on long-term hematopoietic stem cells (LT-HSCs). We found that persistent activation of NRF2 due to Keap1 deficiency did not change the number of LT-HSCs but reduced their quiescence in steady-state hematopoiesis. During hematopoietic regeneration after bone marrow (BM) transplantation, persistent activation of NRF2 reduced the BM reconstitution capacity of LT-HSCs, suggesting that NRF2 reduces the quiescence of LT-HSCs and promotes their differentiation, leading to eventual exhaustion. Transient activation of NRF2 by an electrophilic reagent also promotes the entry of LT-HSCs into the cell cycle. Taken together, our findings show that NRF2 drives the cell cycle entry and differentiation of LT-HSCs at the expense of their quiescence and maintenance, an effect that appears to be beneficial for prompt recovery from blood loss. We propose that the appropriate control of NRF2 activity by KEAP1 is essential for maintaining HSCs and guarantees their stress-induced regenerative response.
Collapse
|
217
|
Morris DC, Cheung WL, Loi R, Zhang T, Lu M, Zhang ZG, Chopp M. Thymosin β4 for the treatment of acute stroke in aged rats. Neurosci Lett 2017; 659:7-13. [PMID: 28864242 DOI: 10.1016/j.neulet.2017.08.064] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/10/2017] [Accepted: 08/28/2017] [Indexed: 12/21/2022]
Abstract
Thymosin β4 (Tβ4) is a 5K peptide which influences cellular migration by inhibiting organization of the actin-cytoskeleton. Tβ4 has neurorestorative properties and is a potential candidate for the treatment of sub-acute stroke. Previous research demonstrated that Tβ4 improved neurological outcome in a young (3 months) rat model of embolic stroke. We hypothesized that Tβ4 would improve neurological outcome in an aged rat model of embolic stroke when administered 24h after embolic stroke. Aged Male Wistar rats (Charles River, France 18-21 months) were subjected to embolic middle cerebral artery occlusion (MCAo). Rats were randomized to receive Tβ4 (12mg/kg, RegeneRx Biopharmaceuticals, Inc.) or control 24h after MCAo and then every 3days for 4 additional doses. The dose of 12mg/kg was the maximal dose of Tβ4 that showed functional improvement in a young rat model of embolic stroke. Functional tests (adhesive-removal test (ART), foot fault test (FFT) and the modified Neurological Severity Score (mNSS)) were performed weekly. The rats were sacrificed 56days after MCAo and lesion volumes were measured. Immunohistochemical analysis for oligodendrogenesis, myelination and gliosis was also performed. Twenty-three rats were included in the study: control group (n=12) and Tβ4 group (n=11). After randomization, there were three deaths in both the control and Tβ4 groups. The Tβ4 treatment reduced infarct volume by more than 50% (12.8%±9.3%, mean±SE, p<0.05) compared to the control group (26.0%±4.3%). However, Tβ4 did not show improvement in functional outcome compared to control. There was no significant increase in oligodendrogenesis, myelination and gliosis between control and treatment with Tβ4, however, we unexpectedly observed that overall (control and Tβ4 groups) astrocytic gliosis as measured by GFAP immunoreactivity was significantly inversely correlated with neurological outcome measured using the modified Neurological Severity Score (mNSS) (p<0.01), suggesting that greater gliosis may be related to improvement of neurological outcome in aged rats. In summary, Tβ4 treatment of stroke aged rats significantly reduces infarct volume compared to vehicle treated stroke, however, Tβ4 treatment did not show improvement in functional outcome, myelination or gliosis when compared to control. GFAP staining was significantly inversely correlated to improvement in the mNSS, suggesting that gliosis in the aged rat may be of benefit in improvement of functional outcome.
Collapse
Affiliation(s)
- Daniel C Morris
- Department of Emergency Medicine, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Wing Lee Cheung
- Department of Emergency Medicine, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Richard Loi
- Department of Emergency Medicine, Henry Ford Health System, Detroit, MI 48202, USA
| | - Talan Zhang
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Mei Lu
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Zheng G Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; Department of Physics, Oakland University, Rochester, MI 48309, USA.
| |
Collapse
|
218
|
Buchwalter A, Hetzer MW. Nucleolar expansion and elevated protein translation in premature aging. Nat Commun 2017; 8:328. [PMID: 28855503 PMCID: PMC5577202 DOI: 10.1038/s41467-017-00322-z] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 06/22/2017] [Indexed: 01/08/2023] Open
Abstract
Premature aging disorders provide an opportunity to study the mechanisms that drive aging. In Hutchinson-Gilford progeria syndrome (HGPS), a mutant form of the nuclear scaffold protein lamin A distorts nuclei and sequesters nuclear proteins. We sought to investigate protein homeostasis in this disease. Here, we report a widespread increase in protein turnover in HGPS-derived cells compared to normal cells. We determine that global protein synthesis is elevated as a consequence of activated nucleoli and enhanced ribosome biogenesis in HGPS-derived fibroblasts. Depleting normal lamin A or inducing mutant lamin A expression are each sufficient to drive nucleolar expansion. We further show that nucleolar size correlates with donor age in primary fibroblasts derived from healthy individuals and that ribosomal RNA production increases with age, indicating that nucleolar size and activity can serve as aging biomarkers. While limiting ribosome biogenesis extends lifespan in several systems, we show that increased ribosome biogenesis and activity are a hallmark of premature aging. HGPS is a premature aging disease caused by mutations in the nuclear protein lamin A. Here, the authors show that cells from patients with HGPS have expanded nucleoli and increased protein synthesis, and report that nucleoli also expand as aging progresses in cells derived from healthy individuals.
Collapse
Affiliation(s)
- Abigail Buchwalter
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Martin W Hetzer
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|
219
|
Kim H, Chang J, Shao L, Han L, Iyer S, Manolagas SC, O'Brien CA, Jilka RL, Zhou D, Almeida M. DNA damage and senescence in osteoprogenitors expressing Osx1 may cause their decrease with age. Aging Cell 2017; 16:693-703. [PMID: 28401730 PMCID: PMC5506444 DOI: 10.1111/acel.12597] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2017] [Indexed: 12/29/2022] Open
Abstract
Age-related bone loss in mice results from a decrease in bone formation and an increase in cortical bone resorption. The former is accounted by a decrease in the number of postmitotic osteoblasts which synthesize the bone matrix and is thought to be the consequence of age-dependent changes in mesenchymal osteoblast progenitors. However, there are no specific markers for these progenitors, and conclusions rely on results from in vitro cultures of mixed cell populations. Moreover, the culprits of such changes remain unknown. Here, we have used Osx1-Cre;TdRFP mice in which osteoprogenitors express the TdRFP fluorescent protein. We report that the number of TdRFP-Osx1 cells, freshly isolated from the bone marrow, declines by more than 50% between 6 and 24 months of age in both female and male mice. Moreover, TdRFP-Osx1 cells from old mice exhibited markers of DNA damage and senescence, such as γH2AX foci, G1 cell cycle arrest, phosphorylation of p53, increased p21CIP1 levels, as well as increased levels of GATA4 and activation of NF-κB - two major stimulators of the senescence-associated secretory phenotype (SASP). Bone marrow stromal cells from old mice also exhibited elevated expression of SASP genes, including several pro-osteoclastogenic cytokines, and increased capacity to support osteoclast formation. These changes were greatly attenuated by the senolytic drug ABT263. Together, these findings suggest that the decline in bone mass with age is the result of intrinsic defects in osteoprogenitor cells, leading to decreased osteoblast numbers and increased support of osteoclast formation.
Collapse
Affiliation(s)
- Ha‐Neui Kim
- Division of Endocrinology and MetabolismCenter for Osteoporosis and Metabolic Bone DiseasesUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Central Arkansas Veterans Healthcare SystemLittle RockARUSA
| | - Jianhui Chang
- Department of Pharmaceutical SciencesUniversity of Arkansas for Medical SciencesLittle RockARUSA
| | - Lijian Shao
- Department of Pharmaceutical SciencesUniversity of Arkansas for Medical SciencesLittle RockARUSA
| | - Li Han
- Division of Endocrinology and MetabolismCenter for Osteoporosis and Metabolic Bone DiseasesUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Central Arkansas Veterans Healthcare SystemLittle RockARUSA
| | - Srividhya Iyer
- Division of Endocrinology and MetabolismCenter for Osteoporosis and Metabolic Bone DiseasesUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Central Arkansas Veterans Healthcare SystemLittle RockARUSA
| | - Stavros C. Manolagas
- Division of Endocrinology and MetabolismCenter for Osteoporosis and Metabolic Bone DiseasesUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Central Arkansas Veterans Healthcare SystemLittle RockARUSA
| | - Charles A. O'Brien
- Division of Endocrinology and MetabolismCenter for Osteoporosis and Metabolic Bone DiseasesUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Central Arkansas Veterans Healthcare SystemLittle RockARUSA
| | - Robert L. Jilka
- Division of Endocrinology and MetabolismCenter for Osteoporosis and Metabolic Bone DiseasesUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Central Arkansas Veterans Healthcare SystemLittle RockARUSA
| | - Daohong Zhou
- Department of Pharmaceutical SciencesUniversity of Arkansas for Medical SciencesLittle RockARUSA
| | - Maria Almeida
- Division of Endocrinology and MetabolismCenter for Osteoporosis and Metabolic Bone DiseasesUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Central Arkansas Veterans Healthcare SystemLittle RockARUSA
| |
Collapse
|
220
|
Sun X, Zhu MJ. AMP-activated protein kinase: a therapeutic target in intestinal diseases. Open Biol 2017; 7:170104. [PMID: 28835570 PMCID: PMC5577448 DOI: 10.1098/rsob.170104] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/25/2017] [Indexed: 02/07/2023] Open
Abstract
Adenosine monophosphate (AMP)-activated protein kinase (AMPK), a highly conserved energy sensor, has a crucial role in cardiovascular, neurodegenerative and inflammatory diseases, as well as in cancer and metabolic disorders. Accumulating studies have demonstrated that AMPK activation enhances paracellular junctions, nutrient transporters, autophagy and apoptosis, and suppresses inflammation and carcinogenesis in the intestine, indicating an essential role of AMPK in intestinal health. AMPK inactivation is an aetiological factor in intestinal dysfunctions. This review summarizes the favourable outcomes of AMPK activation on intestinal health, and discusses AMPK as a potential therapeutic target for intestinal diseases.
Collapse
Affiliation(s)
- Xiaofei Sun
- School of Food Science, Washington State University, Pullman, WA 99164, USA
- School of Food Science, University of Idaho, Moscow, ID 83844, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, USA
- School of Food Science, University of Idaho, Moscow, ID 83844, USA
| |
Collapse
|
221
|
DNA damage tolerance in hematopoietic stem and progenitor cells in mice. Proc Natl Acad Sci U S A 2017; 114:E6875-E6883. [PMID: 28761001 DOI: 10.1073/pnas.1706508114] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
DNA damage tolerance (DDT) enables bypassing of DNA lesions during replication, thereby preventing fork stalling, replication stress, and secondary DNA damage related to fork stalling. Three modes of DDT have been documented: translesion synthesis (TLS), template switching (TS), and repriming. TLS and TS depend on site-specific PCNA K164 monoubiquitination and polyubiquitination, respectively. To investigate the role of DDT in maintaining hematopoietic stem cells (HSCs) and progenitors, we used PcnaK164R/K164R mice as a unique DDT-defective mouse model. Analysis of the composition of HSCs and HSC-derived multipotent progenitors (MPPs) revealed a significantly reduced number of HSCs, likely owing to increased differentiation of HSCs toward myeloid/erythroid-associated MPP2s. This skewing came at the expense of the number of lymphoid-primed MPP4s, which appeared to be compensated for by increased MPP4 proliferation. Furthermore, defective DDT decreased the numbers of MPP-derived common lymphoid progenitor (CLP), common myeloid progenitor (CMP), megakaryocyte-erythroid progenitor (MEP), and granulocyte-macrophage progenitor (GMP) cells, accompanied by increased cell cycle arrest in CMPs. The HSC and MPP phenotypes are reminiscent of premature aging and stressed hematopoiesis, and indeed progressed with age and were exacerbated on cisplatin exposure. Bone marrow transplantations revealed a strong cell intrinsic defect of DDT-deficient HSCs in reconstituting lethally irradiated mice and a strong competitive disadvantage when cotransplanted with wild-type HSCs. These findings indicate a critical role of DDT in maintaining HSCs and progenitor cells, and in preventing premature aging.
Collapse
|
222
|
Li R, Zhang Y, Tu W, Dai Z. Photoelectrochemical Bioanalysis Platform for Cells Monitoring Based on Dual Signal Amplification Using in Situ Generation of Electron Acceptor Coupled with Heterojunction. ACS APPLIED MATERIALS & INTERFACES 2017; 9:22289-22297. [PMID: 28621518 DOI: 10.1021/acsami.7b06107] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
By using in situ generation of electron acceptor coupled with heterojunction as dual signal amplification, a simple photoelectrochemical (PEC) bioanalysis platform was designed. The synergic effect between the photoelectrochemical (PEC) activities of carbon nitride (C3N4) nanosheets and PbS quantum dots (QDs) achieved almost nine-fold photocurrent intensity increment compared with the C3N4 alone. After the G-quadruplex/hemin/Pt nanoparticles (NPs) with catalase-like activity toward H2O2 were introduced, oxygen was in situ generated and acted as electron donor by improving charge separation efficiency and further enhancing photocurrent response. The dually amplified signal made enough sensitivity for monitoring H2O2 released from live cells. The photocathode was prepared by the stepwise assembly of C3N4 nanosheets and PbS QDs on indium tin oxide (ITO) electrode, which was characterized by scanning electron microscope. A signal-on protocol was achieved for H2O2 detection in vitro due to the relevance of photocurrent on the concentration of H2O2. Under the optimized condition, the fabricated PEC bioanalysis platform exhibited a linear range of 10-7000 μM with a detection limit of 1.05 μM at S/N of 3. Besides, the bioanalysis platform displayed good selectivity against other reductive biological species. By using HepG2 cells as a model, a dual signal amplifying PEC bioanalysis platform for monitoring cells was developed. The bioanalysis platform was successfully applied to the detection of H2O2 release from live cells, which provided a novel method for cells monitoring and would have prospect in clinical assay.
Collapse
Affiliation(s)
- Ruyan Li
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials and Jiangsu Key Laboratory of Biofunctional Materials, College of Chemistry and Materials Science, Nanjing Normal University , Nanjing 210023, P. R. China
| | - Yue Zhang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials and Jiangsu Key Laboratory of Biofunctional Materials, College of Chemistry and Materials Science, Nanjing Normal University , Nanjing 210023, P. R. China
| | - Wenwen Tu
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials and Jiangsu Key Laboratory of Biofunctional Materials, College of Chemistry and Materials Science, Nanjing Normal University , Nanjing 210023, P. R. China
| | - Zhihui Dai
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials and Jiangsu Key Laboratory of Biofunctional Materials, College of Chemistry and Materials Science, Nanjing Normal University , Nanjing 210023, P. R. China
| |
Collapse
|
223
|
Storb R, Sandmaier BM. Nonmyeloablative allogeneic hematopoietic cell transplantation. Haematologica 2017; 101:521-30. [PMID: 27132278 DOI: 10.3324/haematol.2015.132860] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 02/05/2016] [Indexed: 11/09/2022] Open
Abstract
Most hematological malignancies occur in older patients. Until recently these patients and those with comorbidities were not candidates for treatment with allogeneic hematopoietic transplantation because they were unable to tolerate the heretofore used high-dose conditioning regimens. The finding that many of the cures achieved with allogeneic hematopoietic transplantation were due to graft-versus-tumor effects led to the development of less toxic and well-tolerated reduced intensity and nonmyeloablative regimens. These regimens enabled allogeneic engraftment, thereby setting the stage for graft-versus-tumor effects. This review summarizes the encouraging early results seen with the new regimens and discusses the two hurdles that need to be overcome for achieving even greater success, disease relapse and graft-versus-host disease.
Collapse
Affiliation(s)
- Rainer Storb
- Fred Hutchinson Cancer Research Center and the University of Washington, Seattle, WA, USA
| | - Brenda M Sandmaier
- Fred Hutchinson Cancer Research Center and the University of Washington, Seattle, WA, USA
| |
Collapse
|
224
|
Sun X, Zhu H, Dong Z, Liu X, Ma X, Han S, Lu F, Wang P, Qian S, Wang C, Shen C, Zhao X, Zou Y, Ge J, Sun A. Mitochondrial aldehyde dehydrogenase-2 deficiency compromises therapeutic effect of ALDH bright cell on peripheral ischemia. Redox Biol 2017; 13:196-206. [PMID: 28582728 PMCID: PMC5458766 DOI: 10.1016/j.redox.2017.05.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 05/16/2017] [Accepted: 05/24/2017] [Indexed: 01/07/2023] Open
Abstract
The autologous ALDH bright (ALDHbr) cell therapy for ischemic injury is clinically safe and effective, while the underlying mechanism remains elusive. Here, we demonstrated that the glycolysis dominant metabolism of ALDHbr cells is permissive to restore blood flow in an ischemic hind limb model compared with bone marrow mononuclear cells (BMNCs). PCR array analysis showed overtly elevated Aldh2 expression of ALDHbr cells following hypoxic challenge. Notably, ALDHbr cells therapy induced blood flow recovery in this model was reduced in case of ALDH2 deficiency. Moreover, significantly reduced glycolysis flux and increased reactive oxygen species (ROS) levels were detected in ALDHbr cell from Aldh2-/- mice. Compromised effect on blood flow recovery was also noticed post transplanting the human ALDHbr cell from ALDH2 deficient patients (GA or AA genotypes) in this ischemic hindlimb mice model. Taken together, our findings illustrate the indispensable role of ALDH2 in maintaining glycolysis dominant metabolism of ALDHbr cell and advocate that patient's Aldh2 genotype is a prerequisite for the efficacy of ALDHbr cell therapy for peripheral ischemia.
Collapse
Affiliation(s)
- Xiaolei Sun
- Institute of Biomedical Science, Fudan University, Shanghai 200032, China
| | - Hong Zhu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai 200032, China
| | - Zhen Dong
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai 200032, China
| | - Xiangwei Liu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai 200032, China
| | - Xin Ma
- Institute of Biomedical Science, Fudan University, Shanghai 200032, China
| | - Shasha Han
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai 200032, China
| | - Fei Lu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai 200032, China
| | - Peng Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai 200032, China
| | - Sanli Qian
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai 200032, China
| | - Cong Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai 200032, China
| | - Cheng Shen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai 200032, China
| | - Xiaona Zhao
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai 200032, China
| | - Yunzeng Zou
- Institute of Biomedical Science, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai 200032, China
| | - Junbo Ge
- Institute of Biomedical Science, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai 200032, China
| | - Aijun Sun
- Institute of Biomedical Science, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai 200032, China.
| |
Collapse
|
225
|
Replenishing exosomes from older bone marrow stromal cells with miR-340 inhibits myeloma-related angiogenesis. Blood Adv 2017; 1:812-823. [PMID: 29296725 DOI: 10.1182/bloodadvances.2016003251] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 04/13/2017] [Indexed: 12/28/2022] Open
Abstract
The study of bone marrow stromal cells (BMSCs) and the exosomes they secrete is considered promising for cancer therapy. However, little is known about the effect of donor age on BMSCs. In the present study, we investigated the therapeutic potential of BMSC exosomes derived from donors of different ages using an in vivo model of hypoxic bone marrow in multiple myeloma (MM). We found that donor age was strongly related to senescent changes in BMSCs. Exosomes derived from young BMSCs significantly inhibited MM-induced angiogenesis in Matrigel plugs. The exosomal microRNA (miRNA) expression profile was different between young and older BMSCs, despite similarities in the size and quantity of exosomes. Of note was the observation that the antiangiogenic effect of older BMSCs was enhanced by direct transfection of miR-340 that was preferentially expressed in exosomes derived from young BMSCs. We found that miR-340 inhibited angiogenesis via the hepatocyte growth factor/c-MET (HGF/c-MET) signaling pathway in endothelial cells. Our data provide new insights into exosome-based cancer therapy by modification of BMSC-derived exosomes.
Collapse
|
226
|
Biechonski S, Yassin M, Milyavsky M. DNA-damage response in hematopoietic stem cells: an evolutionary trade-off between blood regeneration and leukemia suppression. Carcinogenesis 2017; 38:367-377. [PMID: 28334174 DOI: 10.1093/carcin/bgx002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/11/2017] [Indexed: 12/12/2022] Open
Abstract
Self-renewing and multipotent hematopoietic stem cells (HSCs) maintain lifelong hematopoiesis. Their enormous regenerative potential coupled with lifetime persistence in the body, in contrast with the Progenitors, demand tight control of HSCs genome stability. Indeed, failure to accurately repair DNA damage in HSCs is associated with bone marrow failure and accelerated leukemogenesis. Recent observations exposed remarkable differences in several DNA-damage response (DDR) aspects between HSCs and Progenitors, especially in their DNA-repair capacities and susceptibility to apoptosis. Human HSCs in comparison with Progenitors exhibit delayed DNA double-strand break rejoining, persistent DDR signaling activation, higher sensitivity to the cytotoxic effects of ionizing radiation and attenuated expression of DNA-repair genes. Importantly, the distinct DDR of HSCs was also documented in mouse models. Nevertheless, physiological significance and the molecular basis of the HSCs-specific DDR features are only partially understood. Taking radiation-induced DDR as a paradigm, this review will focus on the current advances in understanding the role of cell-intrinsic DDR regulators and the cellular microenvironment in balancing stemness with genome stability. Pre-leukemia HSCs and clonal hematopoiesis evolvement will be discussed as an evolutionary compromise between the need for lifelong blood regeneration and DDR. Uniquely for this review, we outline the differences in HSCs-related DDR as highlighted by various experimental systems and attempt to provide their critical analysis.
Collapse
Affiliation(s)
- Shahar Biechonski
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Muhammad Yassin
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Michael Milyavsky
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| |
Collapse
|
227
|
García-Prat L, Muñoz-Cánoves P. Aging, metabolism and stem cells: Spotlight on muscle stem cells. Mol Cell Endocrinol 2017; 445:109-117. [PMID: 27531569 DOI: 10.1016/j.mce.2016.08.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/11/2016] [Indexed: 12/15/2022]
Abstract
All tissues and organs undergo a progressive regenerative decline as they age. This decline has been mainly attributed to loss of stem cell number and/or function, and both stem cell-intrinsic changes and alterations in local niches and/or systemic environment over time are known to contribute to the stem cell aging phenotype. Advancing in the molecular understanding of the deterioration of stem cell cells with aging is key for targeting the specific causes of tissue regenerative dysfunction at advanced stages of life. Here, we revise exciting recent findings on why stem cells age and the consequences on tissue regeneration, with a special focus on regeneration of skeletal muscle. We also highlight newly identified common molecular pathways affecting diverse types of aging stem cells, such as altered proteostasis, metabolism, or senescence entry, and discuss the questions raised by these findings. Finally, we comment on emerging stem cell rejuvenation strategies, principally emanating from studies on muscle stem cells, which will surely burst tissue regeneration research for future benefit of the increasing human aging population.
Collapse
Affiliation(s)
- Laura García-Prat
- Tissue Regeneration Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra; University (UPF) y CIBERNED, Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Tissue Regeneration Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra; University (UPF) y CIBERNED, Barcelona, Spain; ICREA, Barcelona, Spain.
| |
Collapse
|
228
|
Sertorio M, Du W, Amarachintha S, Wilson A, Pang Q. In Vivo RNAi Screen Unveils PPARγ as a Regulator of Hematopoietic Stem Cell Homeostasis. Stem Cell Reports 2017; 8:1242-1255. [PMID: 28416286 PMCID: PMC5425620 DOI: 10.1016/j.stemcr.2017.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 03/09/2017] [Accepted: 03/10/2017] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem cell (HSC) defects can cause repopulating impairment leading to hematologic diseases. To target HSC deficiency in a disease setting, we exploited the repopulating defect of Fanconi anemia (FA) HSCs to conduct an in vivo short hairpin RNA (shRNA) screen. We exposed Fancd2−/− HSCs to a lentiviral shRNA library targeting 947 genes. We found enrichment of shRNAs targeting genes involved in the PPARγ pathway that has not been linked to HSC homeostasis. PPARγ inhibition by shRNA or chemical compounds significantly improves the repopulating ability of Fancd2−/− HSCs. Conversely, activation of PPARγ in wild-type HSCs impaired hematopoietic repopulation. In mouse HSCs and patient-derived lymphoblasts, PPARγ activation is manifested in upregulating the p53 target p21. PPARγ and co-activators are upregulated in total bone marrow and stem/progenitor cells from FA patients. Collectively, this work illustrates the utility of RNAi technology coupled with HSC transplantation for the discovery of novel genes and pathways involved in stress hematopoiesis. In vivo screening identifies of deleterious Pparγ effect on HSCs Pharmacological activation of Pparγ impaired normal HSC repopulation Inhibition of Pparγ improves Fancd2-deficient HSC repopulation ability
Collapse
Affiliation(s)
- Mathieu Sertorio
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Wei Du
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Surya Amarachintha
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Andrew Wilson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Qishen Pang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
229
|
Zhang L, Sha J, Yang G, Huang X, Bo J, Huang Y. Activation of Notch pathway is linked with epithelial-mesenchymal transition in prostate cancer cells. Cell Cycle 2017; 16:999-1007. [PMID: 28388267 DOI: 10.1080/15384101.2017.1312237] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Notch signaling has been reported to play an essential role in tumorigenesis. Several studies have suggested that Notch receptors could be oncoproteins or tumor suppressors in different types of human cancers. Emerging evidence has suggested that Notch pathway regulates cell growth, apoptosis, cell cycle, and metastasis. In the current study, we explore whether Notch-1 could regulate the cell invasion and migration as well as EMT (epithelial-mesenchymal transition) in prostate cancer cells. We found that overexpression of Notch-1 enhanced cell migration and invasion in PC-3 cells. However, downregulation of Notch-1 retarded cell migration and invasion in prostate cancer cells. Importantly, we observed that overexpression of Notch-1 led to EMT in PC-3 cells. Notably, we found that EMT-type cells are associated with EMT markers change and cancer stem cell phenotype. Taken together, we concluded that downregulation of Notch-1 could be a promising approach for inhibition of invasion in prostate cancer cells, which could be useful for the treatment of metastatic prostate cancer.
Collapse
Affiliation(s)
- Lianhua Zhang
- a Department of Urology , Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Jianjun Sha
- a Department of Urology , Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Guoliang Yang
- a Department of Urology , Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Xuyuan Huang
- a Department of Urology , Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Juanjie Bo
- a Department of Urology , Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Yiran Huang
- a Department of Urology , Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| |
Collapse
|
230
|
Abstract
The FOXO family of transcription factors plays a conserved role in longevity and tissue homeostasis across species. In the mammalian nervous system, emerging evidence has implicated FOXOs in cognitive performance, stem cell maintenance, regeneration, and protection against stress. Much of what we know about neuronal functions of FOXO emerged from recent studies in C. elegans. Similar to mammalian FOXO, the worm FOXO ortholog, called DAF-16, regulates learning and memory, regeneration, and stress resistance in neurons. Here, we discuss the current state of our knowledge of FOXO’s functions in neurons in mammals and invertebrates, and highlight areas where our understanding is limited. Defining the function of FOXO factors in the healthy, aged, and diseased brain may have important implications for improving healthspan and treating neurodegenerative disease.
Collapse
Affiliation(s)
- Sun Y Kim
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Ashley E Webb
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| |
Collapse
|
231
|
Abstract
SIGNIFICANCE In the last years, metabolic reprogramming, fluctuations in bioenergetic fuels, and modulation of oxidative stress became new key hallmarks of tumor development. In cancer, elevated glucose uptake and high glycolytic rate, as a source of adenosine triphosphate, constitute a growth advantage for tumors. This represents the universally known Warburg effect, which gave rise to one major clinical application for detecting cancer cells using glucose analogs: the positron emission tomography scan imaging. Recent Advances: Glucose utilization and carbon sources in tumors are much more heterogeneous than initially thought. Indeed, new studies emerged and revealed a dual capacity of tumor cells for glycolytic and oxidative phosphorylation (OXPHOS) metabolism. OXPHOS metabolism, which relies predominantly on mitochondrial respiration, exhibits fine-tuned regulation of respiratory chain complexes and enhanced antioxidant response or detoxification capacity. CRITICAL ISSUES OXPHOS-dependent cancer cells use alternative oxidizable substrates, such as glutamine and fatty acids. The diversity of carbon substrates fueling neoplastic cells is indicative of metabolic heterogeneity, even within tumors sharing the same clinical diagnosis. Metabolic switch supports cancer cell stemness and their bioenergy-consuming functions, such as proliferation, survival, migration, and invasion. Moreover, reactive oxygen species-induced mitochondrial metabolism and nutrient availability are important for interaction with tumor microenvironment components. Carcinoma-associated fibroblasts and immune cells participate in the metabolic interplay with neoplastic cells. They collectively adapt in a dynamic manner to the metabolic needs of cancer cells, thus participating in tumorigenesis and resistance to treatments. FUTURE DIRECTIONS Characterizing the reciprocal metabolic interplay between stromal, immune, and neoplastic cells will provide a better understanding of treatment resistance. Antioxid. Redox Signal. 26, 462-485.
Collapse
Affiliation(s)
- Géraldine Gentric
- 1 Stress and Cancer Laboratory, Équipe Labelisée LNCC, Institut Curie , Paris, France .,2 Inserm , U830, Paris, France
| | - Virginie Mieulet
- 1 Stress and Cancer Laboratory, Équipe Labelisée LNCC, Institut Curie , Paris, France .,2 Inserm , U830, Paris, France
| | - Fatima Mechta-Grigoriou
- 1 Stress and Cancer Laboratory, Équipe Labelisée LNCC, Institut Curie , Paris, France .,2 Inserm , U830, Paris, France
| |
Collapse
|
232
|
Chen J, Ji X, He Z. Smart Composite Reagent Composed of Double-Stranded DNA-Templated Copper Nanoparticle and SYBR Green I for Hydrogen Peroxide Related Biosensing. Anal Chem 2017; 89:3988-3995. [DOI: 10.1021/acs.analchem.6b04484] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Jinyang Chen
- Key Laboratory of Analytical Chemistry
for Biology and Medicine (Ministry of Education), College
of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Xinghu Ji
- Key Laboratory of Analytical Chemistry
for Biology and Medicine (Ministry of Education), College
of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Zhike He
- Key Laboratory of Analytical Chemistry
for Biology and Medicine (Ministry of Education), College
of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
233
|
Abstract
In this review, Ng and Shyh-Chang review recent metabolomic studies of stem cell metabolism that have revealed how metabolic pathways can convey changes in the extrinsic environment or their niche to program stem cell fates. Advances in metabolomics have deepened our understanding of the roles that specific modes of metabolism play in programming stem cell fates. Here, we review recent metabolomic studies of stem cell metabolism that have revealed how metabolic pathways can convey changes in the extrinsic environment or their niche to program stem cell fates. The metabolic programming of stem cells represents a fine balance between the intrinsic needs of a cellular state and the constraints imposed by extrinsic conditions. A more complete understanding of these needs and constraints will afford us greater mastery over our control of stem cell fates.
Collapse
Affiliation(s)
| | - Huck-Hui Ng
- Genome Institute of Singapore, Singapore 138675
| |
Collapse
|
234
|
PM 2.5 Exposure Suppresses Dendritic Maturation in Subgranular Zone in Aged Rats. Neurotox Res 2017; 32:50-57. [PMID: 28275902 PMCID: PMC5487878 DOI: 10.1007/s12640-017-9710-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/03/2017] [Accepted: 02/13/2017] [Indexed: 01/19/2023]
Abstract
Detrimental effects of long-term inhalation of fine particulate matter (PM2.5) on the pulmonary and cardiovascular systems have been widely reported. Recent studies have shown that exposure to PM2.5 also causes adverse neurocognitive effects. This study investigates the effects of inhaled ammonium sulfate, which is a major compound of inorganic air pollutants in PM2.5, on adult neurogenesis in aged Sprague-Dawley rats. A total of 20 rats were randomly assigned to experimental (n = 10) and control (n = 10) conditions, wherein they were exposed to either ammonium sulfate or sham air for 2 h per day and for 28 consecutive days. It was observed that ammonium sulfate inhibited the maturation process and diminished dendritic complexity of immature neurons in the subgranular zone (SGZ) of the hippocampus significantly, although the number of neural stem cells or the rates of differentiation were comparable between the two groups. Our findings provide clear evidence on the direct relationship between air quality and advantageous neurogenesis. Exposure to PM leads to specific adverse effects on the maturation process during neurogenesis.
Collapse
|
235
|
Ssb1 and Ssb2 cooperate to regulate mouse hematopoietic stem and progenitor cells by resolving replicative stress. Blood 2017; 129:2479-2492. [PMID: 28270450 DOI: 10.1182/blood-2016-06-725093] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 02/26/2017] [Indexed: 12/14/2022] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are vulnerable to endogenous damage and defects in DNA repair can limit their function. The 2 single-stranded DNA (ssDNA) binding proteins SSB1 and SSB2 are crucial regulators of the DNA damage response; however, their overlapping roles during normal physiology are incompletely understood. We generated mice in which both Ssb1 and Ssb2 were constitutively or conditionally deleted. Constitutive Ssb1/Ssb2 double knockout (DKO) caused early embryonic lethality, whereas conditional Ssb1/Ssb2 double knockout (cDKO) in adult mice resulted in acute lethality due to bone marrow failure and intestinal atrophy featuring stem and progenitor cell depletion, a phenotype unexpected from the previously reported single knockout models of Ssb1 or Ssb2 Mechanistically, cDKO HSPCs showed altered replication fork dynamics, massive accumulation of DNA damage, genome-wide double-strand breaks enriched at Ssb-binding regions and CpG islands, together with the accumulation of R-loops and cytosolic ssDNA. Transcriptional profiling of cDKO HSPCs revealed the activation of p53 and interferon (IFN) pathways, which enforced cell cycling in quiescent HSPCs, resulting in their apoptotic death. The rapid cell death phenotype was reproducible in in vitro cultured cDKO-hematopoietic stem cells, which were significantly rescued by nucleotide supplementation or after depletion of p53. Collectively, Ssb1 and Ssb2 control crucial aspects of HSPC function, including proliferation and survival in vivo by resolving replicative stress to maintain genomic stability.
Collapse
|
236
|
Hierarchical tissue organization as a general mechanism to limit the accumulation of somatic mutations. Nat Commun 2017; 8:14545. [PMID: 28230094 PMCID: PMC5331224 DOI: 10.1038/ncomms14545] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 01/11/2017] [Indexed: 01/17/2023] Open
Abstract
How can tissues generate large numbers of cells, yet keep the divisional load (the number of divisions along cell lineages) low in order to curtail the accumulation of somatic mutations and reduce the risk of cancer? To answer the question we consider a general model of hierarchically organized self-renewing tissues and show that the lifetime divisional load of such a tissue is independent of the details of the cell differentiation processes, and depends only on two structural and two dynamical parameters. Our results demonstrate that a strict analytical relationship exists between two seemingly disparate characteristics of self-renewing tissues: divisional load and tissue organization. Most remarkably, we find that a sufficient number of progressively slower dividing cell types can be almost as efficient in minimizing the divisional load, as non-renewing tissues. We argue that one of the main functions of tissue-specific stem cells and differentiation hierarchies is the prevention of cancer. To limit the accumulation of somatic mutations, renewing tissues must minimize the number of times each cell divides during differentiation. Here, the authors analytically derive the lower limit of lifetime divisional load of a tissue, show that hierarchically differentiating tissues can approach this limit, and that this depends on uneven divisional rates across the hierarchy.
Collapse
|
237
|
Hayakawa Y, Fox JG, Wang TC. The Origins of Gastric Cancer From Gastric Stem Cells: Lessons From Mouse Models. Cell Mol Gastroenterol Hepatol 2017; 3:331-338. [PMID: 28462375 PMCID: PMC5404024 DOI: 10.1016/j.jcmgh.2017.01.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/28/2017] [Indexed: 12/24/2022]
Abstract
The cellular origin of digestive cancers has been a long-standing question in the cancer field. Mouse models have identified long-lived stem cells in most organ systems, including the luminal gastrointestinal tract, and numerous studies have pointed to tissue resident stem cells as the main cellular origin of cancer. During gastric carcinogenesis, chronic inflammation induces genetic and epigenetic alterations in long-lived stem cells, along with expansion of stem cell niches, eventually leading to invasive cancer. The gastric corpus and antrum have distinct stem cells and stem cell niches, suggesting differential regulation of cancer initiation at the 2 sites. In this short review, we discuss recent experimental models and human studies, which provide important insights into the pathogenesis of gastric cancer.
Collapse
Affiliation(s)
- Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - James G. Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, New York,Correspondence Address correspondence to: Timothy C. Wang, MD, Division of Digestive and Liver Diseases, Department of Medicine and Irving Cancer Research Center, Columbia University Medical Center, 1130 St Nicholas Avenue, Room 925, New York, New York 10032-3802. fax: (212) 851-4590.Division of Digestive and Liver DiseasesDepartment of Medicine and Irving Cancer Research CenterColumbia University Medical Center1130 St Nicholas AvenueRoom 925New YorkNew York 10032-3802
| |
Collapse
|
238
|
RBM4 Regulates Neuronal Differentiation of Mesenchymal Stem Cells by Modulating Alternative Splicing of Pyruvate Kinase M. Mol Cell Biol 2017; 37:MCB.00466-16. [PMID: 27821480 DOI: 10.1128/mcb.00466-16] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 11/01/2016] [Indexed: 12/31/2022] Open
Abstract
RBM4 promotes differentiation of neuronal progenitor cells and neurite outgrowth of cultured neurons via its role in splicing regulation. In this study, we further explored the role of RBM4 in neuronal differentiation. During neuronal differentiation, energy production shifts from glycolysis to oxidative phosphorylation. We found that the splice isoform change of the metabolic enzyme pyruvate kinase M (PKM) from PKM2 to PKM1 occurs during brain development and is impaired in RBM4-deficient brains. The PKM isoform change could be recapitulated in human mesenchymal stem cells (MSCs) during neuronal induction. Using a PKM minigene, we demonstrated that RBM4 plays a direct role in regulating alternative splicing of PKM. Moreover, RBM4 antagonized the function of the splicing factor PTB and induced the expression of a PTB isoform with attenuated splicing activity in MSCs. Overexpression of RBM4 or PKM1 induced the expression of neuronal genes, increased the mitochondrial respiration capacity in MSCs, and, accordingly, promoted neuronal differentiation. Finally, we demonstrated that RBM4 is induced and is involved in the PKM splicing switch and neuronal gene expression during hypoxia-induced neuronal differentiation. Hence, RBM4 plays an important role in the PKM isoform switch and the change in mitochondrial energy production during neuronal differentiation.
Collapse
|
239
|
Zong C, Wang M, Li B, Liu X, Zhao W, Zhang Q, Liang A, Yu Y. Sensing of hydrogen peroxide and glucose in human serum via quenching fluorescence of biomolecule-stabilized Au nanoclusters assisted by the Fenton reaction. RSC Adv 2017. [DOI: 10.1039/c7ra01498h] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Sensitive detection of H2O2 and glucose were realized by Fenton reaction assistant oxidation of Au NCs.
Collapse
Affiliation(s)
- Chenghua Zong
- Jiangsu Key Laboratory of Green Synthesis for Functional Materials
- School of Chemistry and Material Science
- Jiangsu Normal University
- Xuzhou
- China
| | - Min Wang
- Jiangsu Key Laboratory of Green Synthesis for Functional Materials
- School of Chemistry and Material Science
- Jiangsu Normal University
- Xuzhou
- China
| | - Bo Li
- Jiangsu Key Laboratory of Green Synthesis for Functional Materials
- School of Chemistry and Material Science
- Jiangsu Normal University
- Xuzhou
- China
| | - Xiaojun Liu
- Jiangsu Key Laboratory of Green Synthesis for Functional Materials
- School of Chemistry and Material Science
- Jiangsu Normal University
- Xuzhou
- China
| | - Wenfeng Zhao
- Jiangsu Key Laboratory of Green Synthesis for Functional Materials
- School of Chemistry and Material Science
- Jiangsu Normal University
- Xuzhou
- China
| | - Qingquan Zhang
- Jiangsu Key Laboratory of Green Synthesis for Functional Materials
- School of Chemistry and Material Science
- Jiangsu Normal University
- Xuzhou
- China
| | - Aiye Liang
- Department of Physical Sciences
- Charleston Southern University
- Charleston
- USA
| | - Yang Yu
- Jiangsu Key Laboratory of Green Synthesis for Functional Materials
- School of Chemistry and Material Science
- Jiangsu Normal University
- Xuzhou
- China
| |
Collapse
|
240
|
Elias HK, Bryder D, Park CY. Molecular mechanisms underlying lineage bias in aging hematopoiesis. Semin Hematol 2017; 54:4-11. [DOI: 10.1053/j.seminhematol.2016.11.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
241
|
Hayat M. Overview of Autophagy. AUTOPHAGY: CANCER, OTHER PATHOLOGIES, INFLAMMATION, IMMUNITY, INFECTION, AND AGING 2017:1-122. [DOI: 10.1016/b978-0-12-812146-7.00001-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
242
|
Bigarella CL, Li J, Rimmelé P, Liang R, Sobol RW, Ghaffari S. FOXO3 Transcription Factor Is Essential for Protecting Hematopoietic Stem and Progenitor Cells from Oxidative DNA Damage. J Biol Chem 2016; 292:3005-3015. [PMID: 27994057 DOI: 10.1074/jbc.m116.769455] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 12/16/2016] [Indexed: 12/29/2022] Open
Abstract
Accumulation of damaged DNA in hematopoietic stem cells (HSC) is associated with chromosomal abnormalities, genomic instability, and HSC aging and might promote hematological malignancies with age. Despite this, the regulatory pathways implicated in the HSC DNA damage response have not been fully elucidated. One of the sources of DNA damage is reactive oxygen species (ROS) generated by both exogenous and endogenous insults. Balancing ROS levels in HSC requires FOXO3, which is an essential transcription factor for HSC maintenance implicated in HSC aging. Elevated ROS levels result in defective Foxo3-/- HSC cycling, among many other deficiencies. Here, we show that loss of FOXO3 leads to the accumulation of DNA damage in primitive hematopoietic stem and progenitor cells (HSPC), associated specifically with reduced expression of genes implicated in the repair of oxidative DNA damage. We provide further evidence that Foxo3-/- HSPC are defective in DNA damage repair. Specifically, we show that the base excision repair pathway, the main pathway utilized for the repair of oxidative DNA damage, is compromised in Foxo3-/- primitive hematopoietic cells. Treating mice in vivo with N-acetylcysteine reduces ROS levels, rescues HSC cycling defects, and partially mitigates HSPC DNA damage. These results indicate that DNA damage accrued as a result of elevated ROS in Foxo3-/- mutant HSPC is at least partially reversible. Collectively, our findings suggest that FOXO3 serves as a protector of HSC genomic stability and health.
Collapse
Affiliation(s)
- Carolina L Bigarella
- From the Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Jianfeng Li
- the Department of Oncologic Sciences, University of South Alabama Mitchell Cancer Institute, Mobile, Alabama 36604
| | - Pauline Rimmelé
- From the Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Raymond Liang
- From the Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029.,the Developmental and Stem Cell Biology Multidisciplinary Training Area
| | - Robert W Sobol
- the Department of Oncologic Sciences, University of South Alabama Mitchell Cancer Institute, Mobile, Alabama 36604
| | - Saghi Ghaffari
- From the Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, .,the Developmental and Stem Cell Biology Multidisciplinary Training Area.,Department of Medicine, Division of Hematology and, Oncology.,Black Family Stem Cell Institute, and.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
243
|
Impaired haematopoietic stem cell differentiation and enhanced skewing towards myeloid progenitors in aged caspase-2-deficient mice. Cell Death Dis 2016; 7:e2509. [PMID: 27906175 PMCID: PMC5260989 DOI: 10.1038/cddis.2016.406] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/26/2016] [Accepted: 11/07/2016] [Indexed: 12/24/2022]
Abstract
The apoptotic cysteine protease caspase-2 has been shown to suppress tumourigenesis in mice and its reduced expression correlates with poor prognosis in some human malignancies. Caspase-2-deficient mice develop normally but show ageing-related traits and, when challenged by oncogenic stimuli or certain stress, show enhanced tumour development, often accompanied by extensive aneuploidy. As stem cells are susceptible to acquiring age-related functional defects because of their self-renewal and proliferative capacity, we examined whether loss of caspase-2 promotes such defects with age. Using young and aged Casp2−/− mice, we demonstrate that deficiency of caspase-2 results in enhanced aneuploidy and DNA damage in bone marrow (BM) cells with ageing. Furthermore, we demonstrate for the first time that caspase-2 loss results in significant increase in immunophenotypically defined short-term haematopoietic stem cells (HSCs) and multipotent progenitors fractions in BM with a skewed differentiation towards myeloid progenitors with ageing. Caspase-2 deficiency leads to enhanced granulocyte macrophage and erythroid progenitors in aged mice. Colony-forming assays and long-term culture-initiating assay further recapitulated these results. Our results provide the first evidence of caspase-2 in regulating HSC and progenitor differentiation, as well as aneuploidy, in vivo.
Collapse
|
244
|
Khurana S, Schouteden S, Manesia JK, Santamaria-Martínez A, Huelsken J, Lacy-Hulbert A, Verfaillie CM. Outside-in integrin signalling regulates haematopoietic stem cell function via Periostin-Itgav axis. Nat Commun 2016; 7:13500. [PMID: 27905395 PMCID: PMC5146274 DOI: 10.1038/ncomms13500] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/11/2016] [Indexed: 01/08/2023] Open
Abstract
Integrins play an important role in haematopoietic stem cell (HSC) maintenance in the bone marrow niche. Here, we demonstrate that Periostin (Postn) via interaction with Integrin-αv (Itgav) regulates HSC proliferation. Systemic deletion of Postn results in peripheral blood (PB) anaemia, myelomonocytosis and lymphopenia, while the number of phenotypic HSCs increases in the bone marrow. Postn−/− mice recover faster from radiation injury with concomitant loss of primitive HSCs. HSCs from Postn−/− mice show accumulation of DNA damage generally associated with aged HSCs. Itgav deletion in the haematopoietic system leads to a similar PB phenotype and HSC-intrinsic repopulation defects. Unaffected by Postn, Vav-Itgav−/− HSCs proliferate faster in vitro, illustrating the importance of Postn-Itgav interaction. Finally, the Postn-Itgav interaction inhibits the FAK/PI3K/AKT pathway in HSCs, leading to increase in p27Kip1 expression resulting in improved maintenance of quiescent HSCs. Together, we demonstrate a role for Itgav-mediated outside-in signalling in regulation of HSC proliferation and stemness. Integrins regulate haematopoietic stem cell (HSC) homeostasis and engraftment into the bone marrow (BM) niche upon transplantation. Here, the authors show that HSC quiescence and function in the BM is regulated by the interaction of PERIOSTIN and INTEGRIN αv and subsequent increase in p27Kip1.
Collapse
Affiliation(s)
- Satish Khurana
- Inter-departmental Stem Cell Institute, KU Leuven, 3000 Leuven, Belgium
| | - Sarah Schouteden
- Inter-departmental Stem Cell Institute, KU Leuven, 3000 Leuven, Belgium
| | - Javed K Manesia
- Inter-departmental Stem Cell Institute, KU Leuven, 3000 Leuven, Belgium
| | | | - Joerg Huelsken
- École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Adam Lacy-Hulbert
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | | |
Collapse
|
245
|
Wang Q, Zhang W, He G, Sha H, Quan Z. Method for in vitro differentiation of bone marrow mesenchymal stem cells into endothelial progenitor cells and vascular endothelial cells. Mol Med Rep 2016; 14:5551-5555. [PMID: 27878275 PMCID: PMC5355713 DOI: 10.3892/mmr.2016.5953] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 11/10/2016] [Indexed: 11/26/2022] Open
Abstract
Vascular development is a regulated process and is dependent on the participation and differentiation of many cell types including the proliferation and migration of vascular endothelial cells and differentiation of endothelial progenitor cells (EPCs) to mesodermal precursor cells. Thus, reconstitution of this process in vitro necessitates providing ambient conditions for generating and culturing EPCs in vitro and differentiating them to vascular endothelial cells. In the present study, we developed methods to differentiate bone marrow mesenchymal stem cells (MSC) into EPCs and to vascular endothelial cells. Bone marrow MSC from canines and human sources were differentiated in vitro in to EPCs. These EPCs were able to express a variety of endothelial markers following 7 days in culture. Further culturing led to the appearance of an increased number and proportion of endothelial cells. These cells were stable even after 30 generations in culture. There was a gradual loss of CD31 and increased expression of factor VIII, VEGFR and CD133. VEGF being highly angiogenic, helps in the vascular development. These results provide the basis for the possible development of vasculature in vitro conditions for biomedical applications and in vivo for organ/tissue reconstruction therapies.
Collapse
Affiliation(s)
- Qihong Wang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Weifeng Zhang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Guifen He
- School of Life Science, Fudan University, Shanghai 200433, P.R. China
| | - Huifang Sha
- Chest Tumor Research Institute of Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, P.R. China
| | - Zhe Quan
- Department of Neurosurgery, Fengxian District Central Hospital (Branch Hospital of Shanghai Sixth People's Hospital), Shanghai Jiaotong University, Shanghai 201400, P.R. China
| |
Collapse
|
246
|
Park CY. Hematopoiesis in aging: Current concepts and challenges. Semin Hematol 2016; 54:1-3. [PMID: 28088981 DOI: 10.1053/j.seminhematol.2016.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 10/28/2016] [Indexed: 01/05/2023]
Affiliation(s)
- Christopher Y Park
- Department of Pathology, New York University School of Medicine, New York, NY.
| |
Collapse
|
247
|
Wang Y, Gao C, Ge S, Yu J, Yan M. Platelike WO3 sensitized with CdS quantum dots heterostructures for photoelectrochemical dynamic sensing of H2O2 based on enzymatic etching. Biosens Bioelectron 2016; 85:205-211. [DOI: 10.1016/j.bios.2016.05.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/18/2016] [Accepted: 05/05/2016] [Indexed: 11/24/2022]
|
248
|
Zhao W, Yang S, Yang J, Li J, Zheng J, Qing Z, Yang R. Visual Biopsy by Hydrogen Peroxide-Induced Signal Amplification. Anal Chem 2016; 88:10728-10735. [DOI: 10.1021/acs.analchem.6b03330] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Wenjie Zhao
- State
Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry
and Chemical Engineering, and Collaborative Innovation Center for
Chemistry and Molecular Medicine, Hunan University, Changsha, 410082, People’s Republic of China
| | - Sheng Yang
- School
of Chemistry and Biological Engineering, Changsha University of Science and Technology, Changsha, 410114, People’s Republic of China
| | - Jinfeng Yang
- The
Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410011, People’s Republic of China
| | - Jishan Li
- State
Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry
and Chemical Engineering, and Collaborative Innovation Center for
Chemistry and Molecular Medicine, Hunan University, Changsha, 410082, People’s Republic of China
| | - Jing Zheng
- State
Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry
and Chemical Engineering, and Collaborative Innovation Center for
Chemistry and Molecular Medicine, Hunan University, Changsha, 410082, People’s Republic of China
| | - Zhihe Qing
- School
of Chemistry and Biological Engineering, Changsha University of Science and Technology, Changsha, 410114, People’s Republic of China
| | - Ronghua Yang
- State
Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry
and Chemical Engineering, and Collaborative Innovation Center for
Chemistry and Molecular Medicine, Hunan University, Changsha, 410082, People’s Republic of China
- School
of Chemistry and Biological Engineering, Changsha University of Science and Technology, Changsha, 410114, People’s Republic of China
| |
Collapse
|
249
|
Latchney SE, Calvi LM. The aging hematopoietic stem cell niche: Phenotypic and functional changes and mechanisms that contribute to hematopoietic aging. Semin Hematol 2016; 54:25-32. [PMID: 28088984 DOI: 10.1053/j.seminhematol.2016.10.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 10/13/2016] [Indexed: 11/11/2022]
Abstract
The hematopoietic system has the remarkable ability to provide a lifelong supply of mature cells that make up the entire blood and immune system. However, similar to other adult stem cell niches, the hematopoietic system is vulnerable to the detrimental effects of aging. This is a substantial health concern as the trend for population aging continues to increase. Identifying mechanisms that underlie hematopoietic aging is vital for understanding hematopoietic-related diseases. In this review, we first discuss the cellular hierarchy of the hematopoietic system and the components that make up the surrounding hematopoietic niche. We then provide an overview of the major phenotypes associated with hematopoietic aging and discuss recent research investigating cell-intrinsic and cell-extrinsic mechanisms of hematopoietic stem cell (HSCs) aging. We end by discussing the exciting new concept of possibly reversing the HSC aging process along with outstanding questions that remain to be answered.
Collapse
Affiliation(s)
- Sarah E Latchney
- Endocrine Metabolism Division, University of Rochester School of Medicine and Dentistry, Rochester NY
| | - Laura M Calvi
- Endocrine Metabolism Division, University of Rochester School of Medicine and Dentistry, Rochester NY; Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester NY.
| |
Collapse
|
250
|
Abstract
Cancers induced by gene mutation, deletion, and genome instability might be related to aging. With similar pathways of aging but distinct functions, senescence at the cellular level is an irreversible arrest of cell cycle. Senescence has long been believed as a barrier to restrict tumor expansion. However, more and more evidence has been shown that senescence inducers regulate epithelial-mesenchymal transition, stem cell self-renewal, inflammatory response, crosstalk with the oncogenic bypass signaling, and conversion of oncogene to tumor suppressor. Here we will discuss the most recent findings of the oncogenic aspects of senescence which crosstalk with multiple pathways in cancer progression.
Collapse
Affiliation(s)
- Qing Yang
- a Department of Biology , School of Science and Technology, Nazarbayev University , Astana , Kazakhstan
| | - Yingqiu Xie
- a Department of Biology , School of Science and Technology, Nazarbayev University , Astana , Kazakhstan
| | - Lixia Miao
- b College of Basic Medicine, Wuhan University , Wuhan , China
| |
Collapse
|