251
|
Clémot M, Sênos Demarco R, Jones DL. Lipid Mediated Regulation of Adult Stem Cell Behavior. Front Cell Dev Biol 2020; 8:115. [PMID: 32185173 PMCID: PMC7058546 DOI: 10.3389/fcell.2020.00115] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/11/2020] [Indexed: 12/18/2022] Open
Abstract
Adult stem cells constitute an important reservoir of self-renewing progenitor cells and are crucial for maintaining tissue and organ homeostasis. The capacity of stem cells to self-renew or differentiate can be attributed to distinct metabolic states, and it is now becoming apparent that metabolism plays instructive roles in stem cell fate decisions. Lipids are an extremely vast class of biomolecules, with essential roles in energy homeostasis, membrane structure and signaling. Imbalances in lipid homeostasis can result in lipotoxicity, cell death and diseases, such as cardiovascular disease, insulin resistance and diabetes, autoimmune disorders and cancer. Therefore, understanding how lipid metabolism affects stem cell behavior offers promising perspectives for the development of novel approaches to control stem cell behavior either in vitro or in patients, by modulating lipid metabolic pathways pharmacologically or through diet. In this review, we will first address how recent progress in lipidomics has created new opportunities to uncover stem-cell specific lipidomes. In addition, genetic and/or pharmacological modulation of lipid metabolism have shown the involvement of specific pathways, such as fatty acid oxidation (FAO), in regulating adult stem cell behavior. We will describe and compare findings obtained in multiple stem cell models in order to provide an assessment on whether unique lipid metabolic pathways may commonly regulate stem cell behavior. We will then review characterized and potential molecular mechanisms through which lipids can affect stem cell-specific properties, including self-renewal, differentiation potential or interaction with the niche. Finally, we aim to summarize the current knowledge of how alterations in lipid homeostasis that occur as a consequence of changes in diet, aging or disease can impact stem cells and, consequently, tissue homeostasis and repair.
Collapse
Affiliation(s)
- Marie Clémot
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rafael Sênos Demarco
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - D. Leanne Jones
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
252
|
Luo MJ, Rao SS, Tan YJ, Yin H, Hu XK, Zhang Y, Liu YW, Yue T, Chen LJ, Li L, Huang YR, Qian YX, Liu ZZ, Cao J, Wang ZX, Luo ZW, Wang YY, Xia K, Tang SY, Chen CY, Xie H. Fasting before or after wound injury accelerates wound healing through the activation of pro-angiogenic SMOC1 and SCG2. Am J Cancer Res 2020; 10:3779-3792. [PMID: 32206122 PMCID: PMC7069085 DOI: 10.7150/thno.44115] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 02/05/2020] [Indexed: 12/26/2022] Open
Abstract
Healing of the chronic diabetic ulceration and large burns remains a clinical challenge. Therapeutic fasting has been shown to improve health. Our study tested whether fasting facilitates diabetic and burn wound healing and explored the underlying mechanism. Methods: The effects of fasting on diabetic and burn wound healing were evaluated by analyzing the rates of wound closure, re-epithelialization, scar formation, collagen deposition, skin cell proliferation and neovascularization using histological analyses and immunostaining. In vitro functional assays were conducted to assess fasting and refeeding on the angiogenic activities of endothelial cells. Transcriptome sequencing was employed to identify the differentially expressed genes in endothelial cells after fasting treatment and the role of the candidate genes in the fasting-induced promotion of angiogenesis was demonstrated. Results: Two times of 24-h fasting in a week after but especially before wound injury efficiently induced faster wound closure, better epidermal and dermal regeneration, less scar formation and higher level of angiogenesis in mice with diabetic or burn wounds. In vitro, fasting alone by serum deprivation did not increase, but rather reduced the abilities of endothelial cell to proliferate, migrate and form vessel-like tubes. However, subsequent refeeding did not merely rescue, but further augmented the angiogenic activities of endothelial cells. Transcriptome sequencing revealed that fasting itself, but not the following refeeding, induced a prominent upregulation of a variety of pro-angiogenic genes, including SMOC1 (SPARC related modular calcium binding 1) and SCG2 (secretogranin II). Immunofluorescent staining confirmed the increase of SMOC1 and SCG2 expression in both diabetic and burn wounds after fasting treatment. When the expression of SMOC1 or SCG2 was down-regulated, the fasting/refeeding-induced pro-angiogenic effects were markedly attenuated. Conclusion: This study suggests that fasting combined with refeeding, but not fasting solely, enhance endothelial angiogenesis through the activation of SMOC1 and SCG2, thus facilitating neovascularization and rapid wound healing.
Collapse
|
253
|
Stem and Progenitor Cells in the Pathogenesis and Treatment of Digestive Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:125-157. [PMID: 31898785 DOI: 10.1007/978-3-030-31206-0_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The global epidemic of chronic degenerative diseases expands rapidly. The pathogenesis of these noncommunicable disorders revolves around innate immunity, microbiome, and stem cell alterations. Understanding the mechanisms behind stem cell biology and their regulatory pathways is a key to understanding the origin of human disease. Stem cells are involved in tissue and organ damage and regeneration. The evidence is mounting that not only eukaryotic cells but also gut microbiota may release extracellular microvesicles that are absorbed from the gut into the portal and systemic circulation. Linking the fields of stem cells, innate immunity and microbiome research opens up new avenues to develop novel diagnostic (e.g., biomarkers), therapeutic (e.g., microbiome modulation, stem cell-based medicines), and prognostic (personalized diets) tools. In this chapter, we present the short overview of various stem and progenitor cells of adult tissues circulating in peripheral blood and their role in the pathogenesis and treatment of digestive diseases. We also briefly discuss the role of host-stem cell-microbial interactions as a new frontier of research in gastroenterology.
Collapse
|
254
|
Regulation of Tumor Initiation by the Mitochondrial Pyruvate Carrier. Cell Metab 2020; 31:284-300.e7. [PMID: 31813825 PMCID: PMC7004878 DOI: 10.1016/j.cmet.2019.11.002] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 09/03/2019] [Accepted: 11/05/2019] [Indexed: 01/11/2023]
Abstract
Although metabolic adaptations have been demonstrated to be essential for tumor cell proliferation, the metabolic underpinnings of tumor initiation are poorly understood. We found that the earliest stages of colorectal cancer (CRC) initiation are marked by a glycolytic metabolic signature, including downregulation of the mitochondrial pyruvate carrier (MPC), which couples glycolysis and glucose oxidation through mitochondrial pyruvate import. Genetic studies in Drosophila suggest that this downregulation is required because hyperplasia caused by loss of the Apc or Notch tumor suppressors in intestinal stem cells can be completely blocked by MPC overexpression. Moreover, in two distinct CRC mouse models, loss of Mpc1 prior to a tumorigenic stimulus doubled the frequency of adenoma formation and produced higher grade tumors. MPC loss was associated with a glycolytic metabolic phenotype and increased expression of stem cell markers. These data suggest that changes in cellular pyruvate metabolism are necessary and sufficient to promote cancer initiation.
Collapse
|
255
|
Zhang W, Qu J, Liu GH, Belmonte JCI. The ageing epigenome and its rejuvenation. Nat Rev Mol Cell Biol 2020; 21:137-150. [PMID: 32020082 DOI: 10.1038/s41580-019-0204-5] [Citation(s) in RCA: 287] [Impact Index Per Article: 57.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2019] [Indexed: 02/07/2023]
Abstract
Ageing is characterized by the functional decline of tissues and organs and the increased risk of ageing-associated disorders. Several 'rejuvenating' interventions have been proposed to delay ageing and the onset of age-associated decline and disease to extend healthspan and lifespan. These interventions include metabolic manipulation, partial reprogramming, heterochronic parabiosis, pharmaceutical administration and senescent cell ablation. As the ageing process is associated with altered epigenetic mechanisms of gene regulation, such as DNA methylation, histone modification and chromatin remodelling, and non-coding RNAs, the manipulation of these mechanisms is central to the effectiveness of age-delaying interventions. This Review discusses the epigenetic changes that occur during ageing and the rapidly increasing knowledge of how these epigenetic mechanisms have an effect on healthspan and lifespan extension, and outlines questions to guide future research on interventions to rejuvenate the epigenome and delay ageing processes.
Collapse
Affiliation(s)
- Weiqi Zhang
- Beijing Institute for Brain Disorders, Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China.,Key Laboratory of Genomics and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Jing Qu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Guang-Hui Liu
- Beijing Institute for Brain Disorders, Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China. .,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| | | |
Collapse
|
256
|
Li C, Zhou Y, Rychahou P, Weiss HL, Lee EY, Perry CL, Barrett TA, Wang Q, Evers BM. SIRT2 Contributes to the Regulation of Intestinal Cell Proliferation and Differentiation. Cell Mol Gastroenterol Hepatol 2020; 10:43-57. [PMID: 31954883 PMCID: PMC7210478 DOI: 10.1016/j.jcmgh.2020.01.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Intestinal mucosa undergoes a continual process of proliferation, differentiation, and apoptosis. Disruption of this homeostasis is associated with disorders such as inflammatory bowel disease (IBD). We investigated the role of Sirtuin 2 (SIRT2), a NAD-dependent protein deacetylase, in intestinal epithelial cell (IEC) proliferation and differentiation and the mechanism by which SIRT2 contributes to maintenance of intestinal cell homeostasis. METHODS IECs were collected from SIRT2-deficient mice and patients with IBD. Expression of SIRT2, differentiation markers (mucin2, intestinal alkaline phosphatase, villin, Na,K-ATPase, and lysozyme) and Wnt target genes (EPHB2, AXIN2, and cyclin D1) was determined by western blot, real-time RT-PCR, or immunohistochemical (IHC) staining. IECs were treated with TNF or transfected with siRNA targeting SIRT2. Proliferation was determined by villus height and crypt depth, and Ki67 and cyclin D1 IHC staining. For studies using organoids, intestinal crypts were isolated. RESULTS Increased SIRT2 expression was localized to the more differentiated region of the intestine. In contrast, SIRT2 deficiency impaired proliferation and differentiation and altered stemness in the small intestinal epithelium ex vivo and in vivo. SIRT2-deficient mice showed decreased intestinal enterocyte and goblet cell differentiation but increased the Paneth cell lineage and increased proliferation of IECs. Moreover, we found that SIRT2 inhibits Wnt/β-catenin signaling, which critically regulates IEC proliferation and differentiation. Consistent with a distinct role for SIRT2 in maintenance of gut homeostasis, intestinal mucosa from IBD patients exhibited decreased SIRT2 expression. CONCLUSION We demonstrate that SIRT2, which is decreased in intestinal tissues from IBD patients, regulates Wnt-β-catenin signaling and is important for maintenance of IEC proliferation and differentiation.
Collapse
Affiliation(s)
- Chang Li
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Yuning Zhou
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Piotr Rychahou
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky,Department of Surgery, University of Kentucky, Lexington, Kentucky
| | - Heidi L. Weiss
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Eun Y. Lee
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky,Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky
| | - Courtney L. Perry
- Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Terrence A. Barrett
- Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Qingding Wang
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky,Department of Surgery, University of Kentucky, Lexington, Kentucky,Qingding Wang, PhD, Markey Cancer Center, University of Kentucky, 800 Rose Street, CC140, Lexington, KY 40536-0293. fax: (859) 323-2074.
| | - B. Mark Evers
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky,Department of Surgery, University of Kentucky, Lexington, Kentucky,Correspondence Address correspondence to: B. Mark Evers, MD, Markey Cancer Center, University of Kentucky, 800 Rose Street, CC140, Lexington, KY 40536-0293. fax: (859) 323-2074.
| |
Collapse
|
257
|
He D, Wu H, Xiang J, Ruan X, Peng P, Ruan Y, Chen YG, Wang Y, Yu Q, Zhang H, Habib SL, De Pinho RA, Liu H, Li B. Gut stem cell aging is driven by mTORC1 via a p38 MAPK-p53 pathway. Nat Commun 2020; 11:37. [PMID: 31896747 PMCID: PMC6940394 DOI: 10.1038/s41467-019-13911-x] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/01/2019] [Indexed: 12/25/2022] Open
Abstract
Nutrients are absorbed solely by the intestinal villi. Aging of this organ causes malabsorption and associated illnesses, yet its aging mechanisms remain unclear. Here, we show that aging-caused intestinal villus structural and functional decline is regulated by mTORC1, a sensor of nutrients and growth factors, which is highly activated in intestinal stem and progenitor cells in geriatric mice. These aging phenotypes are recapitulated in intestinal stem cell-specific Tsc1 knockout mice. Mechanistically, mTORC1 activation increases protein synthesis of MKK6 and augments activation of the p38 MAPK-p53 pathway, leading to decreases in the number and activity of intestinal stem cells as well as villus size and density. Targeting p38 MAPK or p53 prevents or rescues ISC and villus aging and nutrient absorption defects. These findings reveal that mTORC1 drives aging by augmenting a prominent stress response pathway in gut stem cells and identify p38 MAPK as an anti-aging target downstream of mTORC1. Intestinal aging is associated with declines in structure and absorption of nutrients. Here, the authors show that aging related intestinal decline is mediated by activation of the mTORC1-p38MAPK-p53 pathway in intestinal stem cells and can be ameliorated by abrogating mTORC1 or p38MAPK activity.
Collapse
Affiliation(s)
- Dan He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hongguang Wu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jinnan Xiang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xinsen Ruan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Peike Peng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Ruan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Ye-Guang Chen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Qiang Yu
- A-STAR Genome Institute of Singapore, Singapore, 138648, Singapore
| | - Hongbing Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Samy L Habib
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX78229, USA
| | - Ronald A De Pinho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China. .,Institute of Traditional Chinese Medicine and Stem Cell Research, School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
258
|
Cheng CW, Yilmaz OH, Mihaylova MM. Strategies for Measuring Induction of Fatty Acid Oxidation in Intestinal Stem and Progenitor Cells. Methods Mol Biol 2020; 2171:53-64. [PMID: 32705635 PMCID: PMC8065345 DOI: 10.1007/978-1-0716-0747-3_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
This protocol describes a multipronged approach that we have created to determine the transcriptional induction of fatty acid oxidation (FAO) genes in Lgr5high intestinal stem cells and a subsequent metabolomics-based approach for assessing fatty acid utilization in the mammalian intestinal crypt. More specifically, we describe methods for crypt isolation followed by a FACS-based purification of stem and progenitor populations and RNA-sequencing analysis. Using this workflow, we can determine both basal gene expression profiles of key metabolic genes as well as corresponding changes in response to altered metabolic states, such as fasting. Subsequently, we describe a complementary metabolomics-based approach that we have developed to assess fatty acid uptake and utilization in the crypt using 13C stable isotope tracing. Combining these approaches, one can gain a better understanding of substrate utilization and the preceding transcriptional changes that accommodate these reactions in physiologic states of low carbohydrate utilization or during overabundance of dietary lipids.
Collapse
Affiliation(s)
- Chia-Wei Cheng
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, Department of Biology, MIT, Cambridge, MA 02139 USA
| | - Omer H. Yilmaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, Department of Biology, MIT, Cambridge, MA 02139 USA,Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA,Departments of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 USA,co-corresponding
| | - Maria M. Mihaylova
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210 USA,co-corresponding
| |
Collapse
|
259
|
Katsyuba E, Romani M, Hofer D, Auwerx J. NAD + homeostasis in health and disease. Nat Metab 2020; 2:9-31. [PMID: 32694684 DOI: 10.1038/s42255-019-0161-5] [Citation(s) in RCA: 369] [Impact Index Per Article: 73.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/12/2019] [Indexed: 12/11/2022]
Abstract
The conceptual evolution of nicotinamide adenine dinucleotide (NAD+) from being seen as a simple metabolic cofactor to a pivotal cosubstrate for proteins regulating metabolism and longevity, including the sirtuin family of protein deacylases, has led to a new wave of scientific interest in NAD+. NAD+ levels decline during ageing, and alterations in NAD+ homeostasis can be found in virtually all age-related diseases, including neurodegeneration, diabetes and cancer. In preclinical settings, various strategies to increase NAD+ levels have shown beneficial effects, thus starting a competitive race to discover marketable NAD+ boosters to improve healthspan and lifespan. Here, we review the basics of NAD+ biochemistry and metabolism, and its roles in health and disease, and we discuss current challenges and the future translational potential of NAD+ research.
Collapse
Affiliation(s)
- Elena Katsyuba
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Nagi Bioscience, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Mario Romani
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Dina Hofer
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Thermo Fisher Scientific, Zug, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
260
|
De Nobrega AK, Luz KV, Lyons LC. Resetting the Aging Clock: Implications for Managing Age-Related Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1260:193-265. [PMID: 32304036 DOI: 10.1007/978-3-030-42667-5_9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Worldwide, individuals are living longer due to medical and scientific advances, increased availability of medical care and changes in public health policies. Consequently, increasing attention has been focused on managing chronic conditions and age-related diseases to ensure healthy aging. The endogenous circadian system regulates molecular, physiological and behavioral rhythms orchestrating functional coordination and processes across tissues and organs. Circadian disruption or desynchronization of circadian oscillators increases disease risk and appears to accelerate aging. Reciprocally, aging weakens circadian function aggravating age-related diseases and pathologies. In this review, we summarize the molecular composition and structural organization of the circadian system in mammals and humans, and evaluate the technological and societal factors contributing to the increasing incidence of circadian disorders. Furthermore, we discuss the adverse effects of circadian dysfunction on aging and longevity and the bidirectional interactions through which aging affects circadian function using examples from mammalian research models and humans. Additionally, we review promising methods for managing healthy aging through behavioral and pharmacological reinforcement of the circadian system. Understanding age-related changes in the circadian clock and minimizing circadian dysfunction may be crucial components to promote healthy aging.
Collapse
Affiliation(s)
- Aliza K De Nobrega
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Kristine V Luz
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Lisa C Lyons
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
261
|
Francescangeli F, De Angelis ML, Zeuner A. Dietary Factors in the Control of Gut Homeostasis, Intestinal Stem Cells, and Colorectal Cancer. Nutrients 2019; 11:nu11122936. [PMID: 31816977 PMCID: PMC6950549 DOI: 10.3390/nu11122936] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/21/2019] [Accepted: 11/27/2019] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is the third commonly diagnosed cancer and the second leading cause of cancer-related deaths worldwide. Global CRC burden is expected to increase by 60% in the next decade, with low-income countries experiencing an escalation of CRC incidence and mortality in parallel to the adoption of western lifestyles. CRC incidence is also sharply increasing in individuals younger than 50 years, often presenting at advanced stages and with aggressive features. Both genetic and environmental factors have been recognized as major contributors for the development of CRC, the latter including diet-related conditions such as chronic inflammation and obesity. In particular, a diet rich in fat and sugars (Western-style diet, WSD) has been shown to induce multiple pathophysiological changes in the intestine linked to an increased risk of CRC. In this scenario, dietary factors have been recently shown to play novel unexpected roles in the regulation of intestinal stem cells (ISCs) and of the gut microbiota, which represent the two main biological systems responsible for intestinal homeostasis. Furthermore, diet is increasingly recognized to play a key role in the neoplastic transformation of ISCs and in the metabolic regulation of colorectal cancer stem cells. This review illustrates novel discoveries on the role of dietary components in regulating intestinal homeostasis and colorectal tumorigenesis. Particular focus is dedicated to new areas of research with potential clinical relevance including the effect of food components on ISCs and cancer stem cells (CSCs), the existence of CRC-specific microbial signatures and the alterations of intestinal homeostasis potentially involved in early-onset CRC. New insights on the role of dietary factors in intestinal regulation will provide new tools not only for the prevention and early diagnosis of CRC but also for improving the effectiveness of current CRC therapies.
Collapse
|
262
|
Lewis SK, Nachun D, Martin MG, Horvath S, Coppola G, Jones DL. DNA Methylation Analysis Validates Organoids as a Viable Model for Studying Human Intestinal Aging. Cell Mol Gastroenterol Hepatol 2019; 9:527-541. [PMID: 31805439 PMCID: PMC7044532 DOI: 10.1016/j.jcmgh.2019.11.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS The epithelia of the intestine and colon turn over rapidly and are maintained by adult stem cells at the base of crypts. Although the small intestine and colon have distinct, well-characterized physiological functions, it remains unclear if there are fundamental regional differences in stem cell behavior or region-dependent degenerative changes during aging. Mesenchyme-free organoids provide useful tools for investigating intestinal stem cell biology in vitro and have started to be used for investigating age-related changes in stem cell function. However, it is unknown whether organoids maintain hallmarks of age in the absence of an aging niche. We tested whether stem cell-enriched organoids preserved the DNA methylation-based aging profiles associated with the tissues and crypts from which they were derived. METHODS To address this, we used standard human methylation arrays and the human epigenetic clock as a biomarker of age to analyze in vitro-derived, 3-dimensional, stem cell-enriched intestinal organoids. RESULTS We found that human stem cell-enriched organoids maintained segmental differences in methylation patterns and that age, as measured by the epigenetic clock, also was maintained in vitro. Surprisingly, we found that stem cell-enriched organoids derived from the small intestine showed striking epigenetic age reduction relative to organoids derived from colon. CONCLUSIONS Our data validate the use of organoids as a model for studying human intestinal aging and introduce methods that can be used when modeling aging or age-onset diseases in vitro.
Collapse
Affiliation(s)
- Sophia K. Lewis
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California,Eli and Edythe Broad Stem Cell Research Center, University of California Los Angeles, Los Angeles, California
| | - Daniel Nachun
- Department of Psychiatry and Semel Institute, University of California Los Angeles, Los Angeles, California
| | - Martin G. Martin
- Eli and Edythe Broad Stem Cell Research Center, University of California Los Angeles, Los Angeles, California,Division of Gastroenterology and Nutrition, Department of Pediatrics, Mattel Children's Hospital and David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Steve Horvath
- Department of Human Genetics, Gonda Research Center, David Geffen School of Medicine, Los Angeles, California
| | - Giovanni Coppola
- Department of Psychiatry and Semel Institute, University of California Los Angeles, Los Angeles, California,Department of Neurology, University of California Los Angeles, Los Angeles, California
| | - D. Leanne Jones
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California,Eli and Edythe Broad Stem Cell Research Center, University of California Los Angeles, Los Angeles, California,Correspondence Address correspondence to: D. Leanne Jones, PhD, Department of Molecular, Cell, and Developmental Biology, Terasaki Life Sciences Building Room 5139, 610 Charles E. Young Drive South, University of California Los Angeles, Los Angeles, California 90095.
| |
Collapse
|
263
|
Wipperman MF, Montrose DC, Gotto AM, Hajjar DP. Mammalian Target of Rapamycin: A Metabolic Rheostat for Regulating Adipose Tissue Function and Cardiovascular Health. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:492-501. [PMID: 30803496 DOI: 10.1016/j.ajpath.2018.11.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 11/03/2018] [Accepted: 11/28/2018] [Indexed: 12/31/2022]
Abstract
The complex relationship between diet and metabolism is an important contributor to cellular metabolism and health. Over the past few decades, a central role for mammalian target of rapamycin (mTOR) in the regulation of multiple cellular processes, including the response to food intake, maintaining homeostasis, and the pathogenesis of disease, has been shown. Herein, we first review our current understanding of the biochemical functions of mTOR and its response to fluctuations in hormone levels, like insulin. Second, we highlight the role of mTOR in lipogenesis, adipogenesis, β-oxidation of lipids, and ketosis of carbohydrates, lipids, and proteins. Special attention is paid to recent advances in mTOR signaling in white versus brown adipose tissues. Finally, we review how mTOR regulates cardiovascular health and disease. Together, these insights define a clearer picture of the connection between mTOR signaling, metabolic health, and disease.
Collapse
Affiliation(s)
- Matthew F Wipperman
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York; Clinical and Translational Science Center, Weill Cornell Medicine, Cornell University, New York
| | - David C Montrose
- Department of Pathology, Stony Brook Medicine, Stony Brook, New York
| | - Antonio M Gotto
- Department of Medicine, Weill Cornell Medicine, Cornell University, New York
| | - David P Hajjar
- Department of Pathology and Biochemistry, Weill Cornell Medicine, Cornell University, New York.
| |
Collapse
|
264
|
Cantor JR. The Rise of Physiologic Media. Trends Cell Biol 2019; 29:854-861. [PMID: 31623927 PMCID: PMC7001851 DOI: 10.1016/j.tcb.2019.08.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/23/2019] [Accepted: 08/28/2019] [Indexed: 12/25/2022]
Abstract
Developed decades ago, traditional culture media were not intended to resemble the metabolic composition of human blood, and indeed poorly do so. Yet, despite what is now a clear recognition that environmental factors influence metabolism, such media remain standard to in vitro studies across virtually all areas of biological research. The recent development of physiologic media, like other efforts designed to address the modeling capacity of cell culture, holds immense potential to improve understanding and interpretation of diverse biological and pharmacological studies.
Collapse
Affiliation(s)
- Jason R Cantor
- Morgridge Institute for Research, Madison, WI 53715, USA; Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA; Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
265
|
Abstract
Regenerative processes that maintain the function of the gastrointestinal (GI) epithelium are critical for health and survival of multicellular organisms. In insects and vertebrates, intestinal stem cells (ISCs) regenerate the GI epithelium. ISC function is regulated by intrinsic, local, and systemic stimuli to adjust regeneration to tissue demands. These control mechanisms decline with age, resulting in significant perturbation of intestinal homeostasis. Processes that lead to this decline have been explored intensively in Drosophila melanogaster in recent years and are now starting to be characterized in mammalian models. This review presents a model for age-related regenerative decline in the fly intestine and discusses recent findings that start to establish molecular mechanisms of age-related decline of mammalian ISC function.
Collapse
Affiliation(s)
- Heinrich Jasper
- Immunology Discovery, Genentech, Inc., South San Francisco, California 94080, USA;
| |
Collapse
|
266
|
Neumann B, Baror R, Zhao C, Segel M, Dietmann S, Rawji KS, Foerster S, McClain CR, Chalut K, van Wijngaarden P, Franklin RJM. Metformin Restores CNS Remyelination Capacity by Rejuvenating Aged Stem Cells. Cell Stem Cell 2019; 25:473-485.e8. [PMID: 31585093 PMCID: PMC6863391 DOI: 10.1016/j.stem.2019.08.015] [Citation(s) in RCA: 282] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/27/2019] [Accepted: 08/23/2019] [Indexed: 01/06/2023]
Abstract
The age-related failure to produce oligodendrocytes from oligodendrocyte progenitor cells (OPCs) is associated with irreversible neurodegeneration in multiple sclerosis (MS). Consequently, regenerative approaches have significant potential for treating chronic demyelinating diseases. Here, we show that the differentiation potential of adult rodent OPCs decreases with age. Aged OPCs become unresponsive to pro-differentiation signals, suggesting intrinsic constraints on therapeutic approaches aimed at enhancing OPC differentiation. This decline in functional capacity is associated with hallmarks of cellular aging, including decreased metabolic function and increased DNA damage. Fasting or treatment with metformin can reverse these changes and restore the regenerative capacity of aged OPCs, improving remyelination in aged animals following focal demyelination. Aged OPCs treated with metformin regain responsiveness to pro-differentiation signals, suggesting synergistic effects of rejuvenation and pro-differentiation therapies. These findings provide insight into aging-associated remyelination failure and suggest therapeutic interventions for reversing such declines in chronic disease.
Collapse
Affiliation(s)
- Björn Neumann
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Roey Baror
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Chao Zhao
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Michael Segel
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Sabine Dietmann
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Khalil S Rawji
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Sarah Foerster
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Crystal R McClain
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Kevin Chalut
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK; Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK
| | - Peter van Wijngaarden
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK; Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia; Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia.
| | - Robin J M Franklin
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK.
| |
Collapse
|
267
|
Coller HA. The paradox of metabolism in quiescent stem cells. FEBS Lett 2019; 593:2817-2839. [PMID: 31531979 DOI: 10.1002/1873-3468.13608] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022]
Abstract
The shift between a proliferating and a nonproliferating state is associated with significant changes in metabolic needs. Proliferating cells tend to have higher metabolic rates, and their metabolic profiles facilitate biosynthesis, as compared to those of nondividing cells of the same sort. Recent studies have elucidated specific molecules that control metabolic changes while cells shift between proliferation and quiescence. Embryonic stem cells, which are rapidly proliferating, tend to have metabolic patterns that are similar to those of nonstem cells in a proliferative state. Moreover, although adult stem cells tend to be quiescent, their metabolic profiles have been reported in multiple organs to more closely resemble those of proliferating than those of nondividing cells in some respects. The findings raise questions about whether there are metabolic profiles that are required for stemness, and whether these profiles relate to the metabolic properties that may be required for quiescence. Here, we review the literature on how metabolism changes upon commitment to proliferation and compare the proliferating and nonproliferating metabolic states of differentiated cells and embryonic and adult stem cells.
Collapse
Affiliation(s)
- Hilary A Coller
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA.,Department of Biological Chemistry, David Geffen School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
268
|
Stine RR, Sakers AP, TeSlaa T, Kissig M, Stine ZE, Kwon CW, Cheng L, Lim HW, Kaestner KH, Rabinowitz JD, Seale P. PRDM16 Maintains Homeostasis of the Intestinal Epithelium by Controlling Region-Specific Metabolism. Cell Stem Cell 2019; 25:830-845.e8. [PMID: 31564549 DOI: 10.1016/j.stem.2019.08.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 06/10/2019] [Accepted: 08/28/2019] [Indexed: 01/18/2023]
Abstract
Metabolic pathways dynamically regulate tissue development and maintenance. However, the mechanisms that govern the metabolic adaptation of stem or progenitor cells to their local niche are poorly understood. Here, we define the transcription factor PRDM16 as a region-specific regulator of intestinal metabolism and epithelial renewal. PRDM16 is selectively expressed in the upper intestine, with enrichment in crypt-resident progenitor cells. Acute Prdm16 deletion in mice triggered progenitor apoptosis, leading to diminished epithelial differentiation and severe intestinal atrophy. Genomic and metabolic analyses showed that PRDM16 transcriptionally controls fatty acid oxidation (FAO) in crypts. Expression of this PRDM16-driven FAO program was highest in the upper small intestine and declined distally. Accordingly, deletion of Prdm16 or inhibition of FAO selectively impaired the development and maintenance of upper intestinal enteroids, and these effects were rescued by acetate treatment. Collectively, these data reveal that regionally specified metabolic programs regulate intestinal maintenance.
Collapse
Affiliation(s)
- Rachel R Stine
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Alexander P Sakers
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tara TeSlaa
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Megan Kissig
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Chan Wook Kwon
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lan Cheng
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hee-Woong Lim
- Department of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Klaus H Kaestner
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity & Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
269
|
Cheng CW, Biton M, Haber AL, Gunduz N, Eng G, Gaynor LT, Tripathi S, Calibasi-Kocal G, Rickelt S, Butty VL, Moreno-Serrano M, Iqbal AM, Bauer-Rowe KE, Imada S, Ulutas MS, Mylonas C, Whary MT, Levine SS, Basbinar Y, Hynes RO, Mino-Kenudson M, Deshpande V, Boyer LA, Fox JG, Terranova C, Rai K, Piwnica-Worms H, Mihaylova MM, Regev A, Yilmaz ÖH. Ketone Body Signaling Mediates Intestinal Stem Cell Homeostasis and Adaptation to Diet. Cell 2019; 178:1115-1131.e15. [PMID: 31442404 PMCID: PMC6732196 DOI: 10.1016/j.cell.2019.07.048] [Citation(s) in RCA: 237] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 06/03/2019] [Accepted: 07/25/2019] [Indexed: 01/18/2023]
Abstract
Little is known about how metabolites couple tissue-specific stem cell function with physiology. Here we show that, in the mammalian small intestine, the expression of Hmgcs2 (3-hydroxy-3-methylglutaryl-CoA synthetase 2), the gene encoding the rate-limiting enzyme in the production of ketone bodies, including beta-hydroxybutyrate (βOHB), distinguishes self-renewing Lgr5+ stem cells (ISCs) from differentiated cell types. Hmgcs2 loss depletes βOHB levels in Lgr5+ ISCs and skews their differentiation toward secretory cell fates, which can be rescued by exogenous βOHB and class I histone deacetylase (HDAC) inhibitor treatment. Mechanistically, βOHB acts by inhibiting HDACs to reinforce Notch signaling, instructing ISC self-renewal and lineage decisions. Notably, although a high-fat ketogenic diet elevates ISC function and post-injury regeneration through βOHB-mediated Notch signaling, a glucose-supplemented diet has the opposite effects. These findings reveal how control of βOHB-activated signaling in ISCs by diet helps to fine-tune stem cell adaptation in homeostasis and injury.
Collapse
Affiliation(s)
- Chia-Wei Cheng
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA
| | - Moshe Biton
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Adam L Haber
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Nuray Gunduz
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA; Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, 06800 Ankara, Turkey
| | - George Eng
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA; Department of Pathology, Massachusetts General Hospital Boston and Harvard Medical School, Boston, MA 02114, USA
| | - Liam T Gaynor
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Surya Tripathi
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA
| | - Gizem Calibasi-Kocal
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA; Dokuz Eylul University, Institute of Oncology, Department of Translational Oncology, Izmir, Turkey
| | - Steffen Rickelt
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA
| | - Vincent L Butty
- BioMicro Center at MIT, Department of Biology, MIT, Cambridge, MA 02139, USA
| | | | - Ameena M Iqbal
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA
| | | | - Shinya Imada
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA; Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Mehmet Sefa Ulutas
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA; Department of Biology, Siirt University, Science and Arts Faculty, 56100 Siirt, Turkey
| | | | - Mark T Whary
- Division of Comparative Medicine, Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | - Stuart S Levine
- BioMicro Center at MIT, Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Yasemin Basbinar
- Dokuz Eylul University, Institute of Oncology, Department of Translational Oncology, Izmir, Turkey
| | - Richard O Hynes
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital Boston and Harvard Medical School, Boston, MA 02114, USA
| | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital Boston and Harvard Medical School, Boston, MA 02114, USA
| | | | - James G Fox
- Division of Comparative Medicine, Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | - Christopher Terranova
- Genomic Medicine Department, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kunal Rai
- Genomic Medicine Department, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Helen Piwnica-Worms
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maria M Mihaylova
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210 USA
| | - Aviv Regev
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA; Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Ömer H Yilmaz
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA; Department of Biology, MIT, Cambridge, MA 02139, USA; Department of Pathology, Massachusetts General Hospital Boston and Harvard Medical School, Boston, MA 02114, USA; Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
270
|
Wu SY, Liang J, Yang BC, Leung PS. SIRT1 Activation Promotes β-Cell Regeneration by Activating Endocrine Progenitor Cells via AMPK Signaling-Mediated Fatty Acid Oxidation. Stem Cells 2019; 37:1416-1428. [PMID: 31400234 DOI: 10.1002/stem.3073] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/04/2019] [Accepted: 07/21/2019] [Indexed: 12/26/2022]
Abstract
Induction of β-cell regeneration from endogenous cells represents a highly promising strategy in stem cell-based treatment for patients with diabetes. Recently, calorie restriction has been shown to affect the regulation of tissue and cell regeneration, including β cells, via metabolic related mechanisms. Here, we examined the potential utility of sirtuin 1 (SIRT1), a calorie restriction mimetic, for stimulating β-cell regeneration and the underlying mechanisms of such stimulation. The present results showed that SIRT1 activation with SRT1720 promoted β-cell regeneration in streptozotocin (STZ)-induced β-cell-deficient neonatal rats. This beneficial effect involved enhanced activation of neurogenin3 (NGN3)-positive endocrine progenitors from pancreatic ductal cells, rather than an expansion of residual β cells. A dynamic expression profile of SIRT1 was observed in endocrine progenitors both during β-cell regeneration in neonatal rats and in the second transition phase of mouse pancreas development. Consistently, SRT1720 treatment upregulated endocrine progenitor differentiation in cultured pancreatic rudiments. Upregulation of NGN3 by SIRT1 activation was through stimulating AMP-activated protein kinase (AMPK) signaling-mediated fatty acid oxidation (FAO) in human pancreatic progenitor cells; AMPK inhibition abolished these effects. The present findings demonstrate a promotional effect of SIRT1 activation on β-cell restoration and endocrine progenitor differentiation that involves regulation of AMPK signaling-mediated FAO. Stem Cells 2019;37:1416-1428.
Collapse
Affiliation(s)
- Shang Ying Wu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Juan Liang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Bao Chen Yang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Po Sing Leung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China.,Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
271
|
Venkateswaran N, Lafita-Navarro MC, Hao YH, Kilgore JA, Perez-Castro L, Braverman J, Borenstein-Auerbach N, Kim M, Lesner NP, Mishra P, Brabletz T, Shay JW, DeBerardinis RJ, Williams NS, Yilmaz OH, Conacci-Sorrell M. MYC promotes tryptophan uptake and metabolism by the kynurenine pathway in colon cancer. Genes Dev 2019; 33:1236-1251. [PMID: 31416966 PMCID: PMC6719621 DOI: 10.1101/gad.327056.119] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/12/2019] [Indexed: 11/24/2022]
Abstract
Tumors display increased uptake and processing of nutrients to fulfill the demands of rapidly proliferating cancer cells. Seminal studies have shown that the proto-oncogene MYC promotes metabolic reprogramming by altering glutamine uptake and metabolism in cancer cells. How MYC regulates the metabolism of other amino acids in cancer is not fully understood. Using high-performance liquid chromatography (HPLC)-tandem mass spectrometry (LC-MS/MS), we found that MYC increased intracellular levels of tryptophan and tryptophan metabolites in the kynurenine pathway. MYC induced the expression of the tryptophan transporters SLC7A5 and SLC1A5 and the enzyme arylformamidase (AFMID), involved in the conversion of tryptophan into kynurenine. SLC7A5, SLC1A5, and AFMID were elevated in colon cancer cells and tissues, and kynurenine was significantly greater in tumor samples than in the respective adjacent normal tissue from patients with colon cancer. Compared with normal human colonic epithelial cells, colon cancer cells were more sensitive to the depletion of tryptophan. Blocking enzymes in the kynurenine pathway caused preferential death of established colon cancer cells and transformed colonic organoids. We found that only kynurenine and no other tryptophan metabolite promotes the nuclear translocation of the transcription factor aryl hydrocarbon receptor (AHR). Blocking the interaction between AHR and kynurenine with CH223191 reduced the proliferation of colon cancer cells. Therefore, we propose that limiting cellular kynurenine or its downstream targets could present a new strategy to reduce the proliferation of MYC-dependent cancer cells.
Collapse
Affiliation(s)
- Niranjan Venkateswaran
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - M Carmen Lafita-Navarro
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Yi-Heng Hao
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Jessica A Kilgore
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Lizbeth Perez-Castro
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Jonathan Braverman
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Nofit Borenstein-Auerbach
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Min Kim
- Lydia Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Nicholas P Lesner
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Prashant Mishra
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Thomas Brabletz
- Nikolaus-Fiebiger-Center for Molecular Medicine, University Erlangen-Nurnberg, Erlangen 91054, Germany
| | - Jerry W Shay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Howard Hughes Medical Institute, Dallas, Texas 75390, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Omer H Yilmaz
- Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.,Department of Pathology, Massachusetts General Hospital Boston, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Maralice Conacci-Sorrell
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
272
|
DNA damage in aging, the stem cell perspective. Hum Genet 2019; 139:309-331. [PMID: 31324975 DOI: 10.1007/s00439-019-02047-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 07/05/2019] [Indexed: 02/07/2023]
Abstract
DNA damage is one of the most consistent cellular process proposed to contribute to aging. The maintenance of genomic and epigenomic integrity is critical for proper function of cells and tissues throughout life, and this homeostasis is under constant strain from both extrinsic and intrinsic insults. Considering the relationship between lifespan and genotoxic burden, it is plausible that the longest-lived cellular populations would face an accumulation of DNA damage over time. Tissue-specific stem cells are multipotent populations residing in localized niches and are responsible for maintaining all lineages of their resident tissue/system throughout life. However, many of these stem cells are impacted by genotoxic stress. Several factors may dictate the specific stem cell population response to DNA damage, including the niche location, life history, and fate decisions after damage accrual. This leads to differential handling of DNA damage in different stem cell compartments. Given the importance of adult stem cells in preserving normal tissue function during an individual's lifetime, DNA damage sensitivity and accumulation in these compartments could have crucial implications for aging. Despite this, more support for direct functional effects driven by accumulated DNA damage in adult stem cell compartments is needed. This review will present current evidence for the accumulation and potential influence of DNA damage in adult tissue-specific stem cells and propose inquiry directions that could benefit individual healthspan.
Collapse
|
273
|
Sihag J, Jones PJH. Dietary fatty acid profile influences circulating and tissue fatty acid ethanolamide concentrations in a tissue-specific manner in male Syrian hamsters. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1563-1579. [PMID: 31301433 DOI: 10.1016/j.bbalip.2019.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/04/2019] [Accepted: 07/07/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND The discovery of N‑acylethanolamines (NAEs) has prompted an increase in research aimed at understanding their biological roles including regulation of appetite and energy metabolism. However, a knowledge gap remains to understand the effect of dietary components on NAE levels, in particular, heterogeneity in dietary fatty acid (DFA) profile, on NAE levels across various organs. OBJECTIVE To identify and elucidate the impact of diet on NAE levels in seven different tissues/organs of male hamsters, with the hypothesis that DFA will act as precursors for NAE synthesis in golden Syrian male hamsters. METHOD A two-month feeding trial was performed, wherein hamsters were fed various dietary oil blends with different composition of 18-C fatty acid (FA). RESULTS DFA directly influences tissue FA and NAE levels. After C18:1n9-enriched dietary treatments, marked increases were observed in duodenal C18:1n9 and oleoylethanolamide (OEA) concentrations. Among all tissues; adipose tissue brown, adipose tissue white, brain, heart, intestine-duodenum, intestine-jejunum, and liver, a negative correlation was observed between gut-brain OEA concentrations and body weight. CONCLUSION DFA composition influences FA and NAE levels across all tissues, leading to significant shifts in intestinal-brain OEA concentrations. The endogenously synthesized increased OEA levels in these tissues enable the gut-brain-interrelationship. Henceforth, we summarize that the brain transmits anorexic properties mediated via neuronal signalling, which may contribute to the maintenance of healthy body weight. Thus, the benefits of OEA can be enhanced by the inclusion of C18:1n9-enriched diets, pointing to the possible nutritional use of this naturally occurring bioactive lipid-amide in the management of obesity.
Collapse
Affiliation(s)
- Jyoti Sihag
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, Winnipeg, Manitoba, Canada
| | - Peter J H Jones
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
274
|
Pentinmikko N, Iqbal S, Mana M, Andersson S, Cognetta AB, Suciu RM, Roper J, Luopajärvi K, Markelin E, Gopalakrishnan S, Smolander OP, Naranjo S, Saarinen T, Juuti A, Pietiläinen K, Auvinen P, Ristimäki A, Gupta N, Tammela T, Jacks T, Sabatini DM, Cravatt BF, Yilmaz ÖH, Katajisto P. Notum produced by Paneth cells attenuates regeneration of aged intestinal epithelium. Nature 2019; 571:398-402. [PMID: 31292548 DOI: 10.1038/s41586-019-1383-0] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/10/2019] [Indexed: 12/12/2022]
Abstract
A decline in stem cell function impairs tissue regeneration during ageing, but the role of the stem-cell-supporting niche in ageing is not well understood. The small intestine is maintained by actively cycling intestinal stem cells that are regulated by the Paneth cell niche1,2. Here we show that the regenerative potential of human and mouse intestinal epithelium diminishes with age owing to defects in both stem cells and their niche. The functional decline was caused by a decrease in stemness-maintaining Wnt signalling due to production of Notum, an extracellular Wnt inhibitor, in aged Paneth cells. Mechanistically, high activity of mammalian target of rapamycin complex 1 (mTORC1) in aged Paneth cells inhibits activity of peroxisome proliferator activated receptor α (PPAR-α)3, and lowered PPAR-α activity increased Notum expression. Genetic targeting of Notum or Wnt supplementation restored function of aged intestinal organoids. Moreover, pharmacological inhibition of Notum in mice enhanced the regenerative capacity of aged stem cells and promoted recovery from chemotherapy-induced damage. Our results reveal a role of the stem cell niche in ageing and demonstrate that targeting of Notum can promote regeneration of aged tissues.
Collapse
Affiliation(s)
- Nalle Pentinmikko
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sharif Iqbal
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Miyeko Mana
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA, USA
| | - Simon Andersson
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Armand B Cognetta
- The Skaggs Institute for Chemical Biology, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Radu M Suciu
- The Skaggs Institute for Chemical Biology, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Jatin Roper
- Department of Medicine, Division of Gastroenterology, Duke University, Durham, NC, USA
| | - Kalle Luopajärvi
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Eino Markelin
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | | | - Santiago Naranjo
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA, USA
| | - Tuure Saarinen
- Obesity Research Unit, Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland.,Abdominal Center, Department of Gastrointestinal Surgery, Helsinki University Hospital, Helsinki, Finland
| | - Anne Juuti
- Abdominal Center, Department of Gastrointestinal Surgery, Helsinki University Hospital, Helsinki, Finland
| | - Kirsi Pietiläinen
- Obesity Research Unit, Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Petri Auvinen
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ari Ristimäki
- Department of Pathology, Research Programs Unit and HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Nitin Gupta
- Atlanta Gastroenterology Associates, Atlanta, GA, USA
| | - Tuomas Tammela
- Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tyler Jacks
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA, USA.,Howard Hughes Medical Institute, MIT, Cambridge, MA, USA
| | - David M Sabatini
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA, USA.,Howard Hughes Medical Institute, MIT, Cambridge, MA, USA.,Whitehead Institute for Biomedical Research, Howard Hughes Medical Institute, Department of Biology, MIT, Cambridge, MA, USA
| | - Benjamin F Cravatt
- The Skaggs Institute for Chemical Biology, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ömer H Yilmaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, MIT, Cambridge, MA, USA
| | - Pekka Katajisto
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland. .,Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland. .,Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
275
|
Igarashi M, Miura M, Williams E, Jaksch F, Kadowaki T, Yamauchi T, Guarente L. NAD + supplementation rejuvenates aged gut adult stem cells. Aging Cell 2019; 18:e12935. [PMID: 30917412 PMCID: PMC6516145 DOI: 10.1111/acel.12935] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 01/05/2019] [Accepted: 02/03/2019] [Indexed: 01/25/2023] Open
Abstract
The tissue decline due to aging is associated with the deterioration of adult stem cell function. Here we show the number and proliferative activity of intestinal stem cells (ISCs) but not Paneth cells decline during aging, as does ISC function assessed ex vivo. Levels of SIRT1 and activity of mTORC1 also decline with aging. The treatment with the NAD(+) precursor nicotinamide riboside (NR) rejuvenates ISCs from aged mice and reverses an impaired ability to repair gut damage. The effect of NR is blocked by the mTORC1 inhibitor rapamycin or the SIRT1 inhibitor EX527. These findings demonstrate that small molecules affecting the NAD/SIRT1/mTORC1 axis may guide a translational path for maintenance of the intestine during aging.
Collapse
Affiliation(s)
- Masaki Igarashi
- Department of Diabetes & Metabolic Diseases, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Masaomi Miura
- Department of Diabetes & Metabolic Diseases, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Eric Williams
- Department of Biology, Glenn Labs for the Science of Aging, and Koch InstituteMITCambridgeMassachusetts
| | | | - Takashi Kadowaki
- Department of Diabetes & Metabolic Diseases, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Toshimasa Yamauchi
- Department of Diabetes & Metabolic Diseases, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Leonard Guarente
- Department of Biology, Glenn Labs for the Science of Aging, and Koch InstituteMITCambridgeMassachusetts
| |
Collapse
|
276
|
Zheng CX, Sui BD, Qiu XY, Hu CH, Jin Y. Mitochondrial Regulation of Stem Cells in Bone Homeostasis. Trends Mol Med 2019; 26:89-104. [PMID: 31126872 DOI: 10.1016/j.molmed.2019.04.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/10/2019] [Accepted: 04/19/2019] [Indexed: 12/13/2022]
Abstract
Mitochondria have emerged as key contributors to the organismal homeostasis, in which mitochondrial regulation of stem cells is becoming increasingly important. Originated from mesenchymal stem cell (MSC) and hematopoietic stem cell (HSC) lineage commitments and interactions, bone is a representative organ where the mitochondrial essentiality to stem cell function has most recently been discovered, underlying skeletal health, aging, and diseases. Furthermore, mitochondrial medications based on modulating stem cell specification are emerging to provide promising therapies to counteract bone aging and pathologies. Here we review the cutting-edge knowledge regarding mitochondrial regulation of stem cells in bone homeostasis, highlighting mechanistic insights as well as mitochondrial strategies for augmented bone healing and tissue regeneration.
Collapse
Affiliation(s)
- Chen-Xi Zheng
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi' an, Shaanxi 710032, China
| | - Bing-Dong Sui
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi' an, Shaanxi 710032, China
| | - Xin-Yu Qiu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi' an, Shaanxi 710032, China
| | - Cheng-Hu Hu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi' an, Shaanxi 710032, China; Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi' an, Shaanxi 710032, China.
| |
Collapse
|
277
|
Armutcu F. Fasting may be an alternative treatment method recommended by physicians. ELECTRONIC JOURNAL OF GENERAL MEDICINE 2019. [DOI: 10.29333/ejgm/104620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
278
|
Abstract
In this issue of Cell Stem Cell, Mihaylova et al. (2018) show that short-term fasting increases serum levels of long-chain fatty acids, presumably derived from the host. This effect in turn can rescue age-related phenotypes that occur in intestinal epithelial stem cells.
Collapse
Affiliation(s)
- Derek A G Barisas
- Department of Pathology and Immunology, Washington University Medical School, Saint Louis, MO 63110, USA
| | - Thaddeus S Stappenbeck
- Department of Pathology and Immunology, Washington University Medical School, Saint Louis, MO 63110, USA.
| |
Collapse
|
279
|
Rangan P, Choi I, Wei M, Navarrete G, Guen E, Brandhorst S, Enyati N, Pasia G, Maesincee D, Ocon V, Abdulridha M, Longo VD. Fasting-Mimicking Diet Modulates Microbiota and Promotes Intestinal Regeneration to Reduce Inflammatory Bowel Disease Pathology. Cell Rep 2019; 26:2704-2719.e6. [PMID: 30840892 PMCID: PMC6528490 DOI: 10.1016/j.celrep.2019.02.019] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/01/2019] [Accepted: 02/06/2019] [Indexed: 12/31/2022] Open
Abstract
Dietary interventions are potentially effective therapies for inflammatory bowel diseases (IBDs). We tested the effect of 4-day fasting-mimicking diet (FMD) cycles on a chronic dextran sodium sulfate (DSS)-induced murine model resulting in symptoms and pathology associated with IBD. These FMD cycles reduced intestinal inflammation, increased stem cell number, stimulated protective gut microbiota, and reversed intestinal pathology caused by DSS, whereas water-only fasting increased regenerative and reduced inflammatory markers without reversing pathology. Transplants of Lactobacillus or fecal microbiota from DSS- and FMD-treated mice reversed DSS-induced colon shortening, reduced inflammation, and increased colonic stem cells. In a clinical trial, three FMD cycles reduced markers associated with systemic inflammation. The effect of FMD cycles on microbiota composition, immune cell profile, intestinal stem cell levels and the reversal of pathology associated with IBD in mice, and the anti-inflammatory effects demonstrated in a clinical trial show promise for FMD cycles to ameliorate IBD-associated inflammation in humans.
Collapse
Affiliation(s)
- Priya Rangan
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Inyoung Choi
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Min Wei
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Gerardo Navarrete
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Esra Guen
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Sebastian Brandhorst
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Nobel Enyati
- USC Dornsife College of Letters, Arts & Sciences, Department of Biological Sciences, University of Southern California, 3551 Trousdale Pkwy, Los Angeles, CA 90089-0191, USA
| | - Gab Pasia
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Daral Maesincee
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Vanessa Ocon
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Maya Abdulridha
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Valter D Longo
- USC Dornsife College of Letters, Arts & Sciences, Department of Biological Sciences, University of Southern California, 3551 Trousdale Pkwy, Los Angeles, CA 90089-0191, USA; Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, 1425 San Pablo St, Los Angeles, CA 90033, USA; IFOM FIRC Institute of Molecular Oncology, Via Adamello 16, Milano 20139, Italy.
| |
Collapse
|
280
|
Zwick RK, Ohlstein B, Klein OD. Intestinal renewal across the animal kingdom: comparing stem cell activity in mouse and Drosophila. Am J Physiol Gastrointest Liver Physiol 2019; 316:G313-G322. [PMID: 30543448 PMCID: PMC6415738 DOI: 10.1152/ajpgi.00353.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The gastrointestinal (GI) tract renews frequently to sustain nutrient digestion and absorption in the face of consistent tissue stress. In many species, proliferative intestinal stem cells (ISCs) are responsible for the repair of the damage arising from chemical and mechanical aspects of food breakdown and exposure to pathogens. As the cellular source of all mature cell types of the intestinal epithelium throughout adulthood, ISCs hold tremendous therapeutic potential for understanding and treating GI disease in humans. This review focuses on recent advances in our understanding of ISC identity, behavior, and regulation during homeostasis and injury-induced repair, as revealed by two major animal models used to study regeneration of the small intestine: Drosophila melanogaster and Mus musculus. We emphasize recent findings from Drosophila that are likely to translate to the mammalian GI system, as well as challenging topics in mouse ISC biology that may be ideally suited for investigation in flies. For context, we begin by reviewing major physiological similarities and distinctions between the Drosophila midgut and mouse small intestine.
Collapse
Affiliation(s)
- Rachel K. Zwick
- 1Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, California
| | - Benjamin Ohlstein
- 2Department of Genetics and Development, Columbia University Medical Center, New York, New York
| | - Ophir D. Klein
- 1Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, California,3Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, California
| |
Collapse
|
281
|
Papsdorf K, Brunet A. Linking Lipid Metabolism to Chromatin Regulation in Aging. Trends Cell Biol 2019; 29:97-116. [PMID: 30316636 PMCID: PMC6340780 DOI: 10.1016/j.tcb.2018.09.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/13/2022]
Abstract
The lifespan of an organism is strongly influenced by environmental factors (including diet) and by internal factors (notably reproductive status). Lipid metabolism is critical for adaptation to external conditions or reproduction. Interestingly, specific lipid profiles are associated with longevity, and increased uptake of certain lipids extends longevity in Caenorhabditis elegans and ameliorates disease phenotypes in humans. How lipids impact longevity, and how lipid metabolism is regulated during aging, is just beginning to be unraveled. This review describes recent advances in the regulation and role of lipids in longevity, focusing on the interaction between lipid metabolism and chromatin states in aging and age-related diseases.
Collapse
Affiliation(s)
- Katharina Papsdorf
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA; Glenn Laboratories for the Biology of Aging, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
282
|
Bland JS. Fasting Physiology and Therapeutic Diets: A Look Back to the Future. Integr Med (Encinitas) 2019; 18:16-21. [PMID: 31341428 PMCID: PMC6601432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The evidence presented at this event demonstrated the multiple clinical benefits of fasting physiology and points toward a future in which the clinical applications of dietary approaches will be well understood and successfully utilized. The conference reflected the scope and breadth of current research efforts in this important clinical area. Clearly, the application of the important new concepts related to fasting physiology that are emerging will require the advocacy and participation of professionals who are well trained in the fields of clinical nutrition and personalized lifestyle medicine.
Collapse
|
283
|
Inhibition of Stearoyl-CoA Desaturase-1 Activity Suppressed SREBP Signaling in Colon Cancer Cells and Their Spheroid Growth. GASTROINTESTINAL DISORDERS 2019. [DOI: 10.3390/gidisord1010014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Unsaturated fatty acids are critical in promoting colon tumorigenesis and its stemness. Stearoyl-CoA desaturase-1 (SCD1) is a rate-limiting lipid desaturase associated with colon cancer cell proliferation and metastasis control. This study aims to evaluate the effects of SCD1 inhibition on colon cancer spheroid growth in a three-dimensional cell culture system. An analysis of clinical data showed that increased SCD1 gene expression in colon tumors was negatively correlated with the prognosis. A chemical inhibitor of SCD1, CAY10566, inhibited the growth of colon cancer cells in both monolayer and sphere cultures. In addition, oleic acid administration—a monounsaturated fatty acid generated by the action of SCD1—prevented the suppression of sphere formation by CAY10566. RNA-sequencing data from 382 colon tumor patient samples obtained from the Cancer Genome Atlas database showed that 806 genes were SCD1-associated genes in human colon cancer. Correlation analysis identified the master regulator of lipid homeostasis sterol regulatory element-binding protein 2 (SREBP2) as a prominent transcription factor, whose expression was positively correlated with SCD1 in human colon cancer. SCD1 knockdown using siRNA in colon cancer samples, suppressed SREBP2 gene expression, providing direct evidence that SREBP signaling is under the control of SCD1 in these cells. Pathway analysis in the Ingenuity Pathways Analysis platform showed that SCD1 expression positively correlated with genes involved in multiple pathways, including lipid synthesis and incorporation, cell proliferation, and tissue tumorigenesis. Further network analysis revealed a central role for Myc in the network hierarchy of the SCD1-correlated genes. These findings suggested that SCD1 inhibition would be an effective strategy for suppressing colon cancer spheroid growth, partly through downregulating SREBP-mediated lipid and cholesterol metabolism and Myc signaling.
Collapse
|
284
|
Abstract
Intestinal homeostasis and regeneration are driven by intestinal stem cells (ISCs) lying in the crypt. In addition to the actively cycling ISCs that maintain daily homeostasis, accumulating evidence supports the existence of other pools of stem/progenitor cells with the capacity to repair damaged tissue and facilitate rapid restoration of intestinal integrity after injuries. Appropriate control of ISCs and other populations of intestinal epithelial cells with stem cell activity is essential for intestinal homeostasis and regeneration while their deregulation is implicated in colorectal tumorigenesis. In this review, we will summarize the recent findings about ISC identity and cellular plasticity in intestine, discuss regulatory mechanisms that control ISCs for intestinal homeostasis and regeneration, and put a particular emphasis on extrinsic niche-derived signaling and intrinsic epigenetic regulation. Moreover, we highlight several fundamental questions about the precise mechanisms conferring robust capacity for intestine to maintain physiological homeostasis and repair injuries.
Collapse
Affiliation(s)
- Deqing Hu
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics; Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Heping, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Heping, Tianjin, China
| | - Han Yan
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics; Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Heping, Tianjin, China
| | - Xi C He
- Stowers Institute for Medical Research, Kansas City, USA
| | - Linheng Li
- Stowers Institute for Medical Research, Kansas City, USA.,Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, China
| |
Collapse
|
285
|
Abstract
The intestinal epithelium is one the fastest renewing tissues in mammals and is endowed with extensive adaptability. The more traditional view of a hierarchical organization of the gut has recently given way to a more dynamic model in which various cell types within the intestinal epithelium can de-differentiate and function as an alternative source of stem cells upon tissue damage and stress conditions such as inflammation and tumorigenesis. Here, we will review the mechanistic principles and key players involved in intestinal plasticity and discuss potential therapeutic implications of cellular plasticity in regenerative medicine and cancer.
Collapse
|
286
|
Shetty AK, Kodali M, Upadhya R, Madhu LN. Emerging Anti-Aging Strategies - Scientific Basis and Efficacy. Aging Dis 2018; 9:1165-1184. [PMID: 30574426 PMCID: PMC6284760 DOI: 10.14336/ad.2018.1026] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022] Open
Abstract
The prevalence of age-related diseases is in an upward trend due to increased life expectancy in humans. Age-related conditions are among the leading causes of morbidity and death worldwide currently. Therefore, there is an urgent need to find apt interventions that slow down aging and reduce or postpone the incidence of debilitating age-related diseases. This review discusses the efficacy of emerging anti-aging approaches for maintaining better health in old age. There are many anti-aging strategies in development, which include procedures such as augmentation of autophagy, elimination of senescent cells, transfusion of plasma from young blood, intermittent fasting, enhancement of adult neurogenesis, physical exercise, antioxidant intake, and stem cell therapy. Multiple pre-clinical studies suggest that administration of autophagy enhancers, senolytic drugs, plasma from young blood, drugs that enhance neurogenesis and BDNF are promising approaches to sustain normal health during aging and also to postpone age-related neurodegenerative diseases such as Alzheimer's disease. Stem cell therapy has also shown promise for improving regeneration and function of the aged or Alzheimer's disease brain. Several of these approaches are awaiting critical appraisal in clinical trials to determine their long-term efficacy and possible adverse effects. On the other hand, procedures such as intermittent fasting, physical exercise, intake of antioxidants such as resveratrol and curcumin have shown considerable promise for improving function in aging, some of which are ready for large-scale clinical trials, as they are non-invasive, and seem to have minimal side effects. In summary, several approaches are at the forefront of becoming mainstream therapies for combating aging and postponing age-related diseases in the coming years.
Collapse
Affiliation(s)
- Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Leelavathi N. Madhu
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
| |
Collapse
|
287
|
Liu Y, Colby JK, Zuo X, Jaoude J, Wei D, Shureiqi I. The Role of PPAR-δ in Metabolism, Inflammation, and Cancer: Many Characters of a Critical Transcription Factor. Int J Mol Sci 2018; 19:3339. [PMID: 30373124 PMCID: PMC6275063 DOI: 10.3390/ijms19113339] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023] Open
Abstract
Peroxisome proliferator-activated receptor-delta (PPAR-δ), one of three members of the PPAR group in the nuclear receptor superfamily, is a ligand-activated transcription factor. PPAR-δ regulates important cellular metabolic functions that contribute to maintaining energy balance. PPAR-δ is especially important in regulating fatty acid uptake, transport, and β-oxidation as well as insulin secretion and sensitivity. These salutary PPAR-δ functions in normal cells are thought to protect against metabolic-syndrome-related diseases, such as obesity, dyslipidemia, insulin resistance/type 2 diabetes, hepatosteatosis, and atherosclerosis. Given the high clinical burden these diseases pose, highly selective synthetic activating ligands of PPAR-δ were developed as potential preventive/therapeutic agents. Some of these compounds showed some efficacy in clinical trials focused on metabolic-syndrome-related conditions. However, the clinical development of PPAR-δ agonists was halted because various lines of evidence demonstrated that cancer cells upregulated PPAR-δ expression/activity as a defense mechanism against nutritional deprivation and energy stresses, improving their survival and promoting cancer progression. This review discusses the complex relationship between PPAR-δ in health and disease and highlights our current knowledge regarding the different roles that PPAR-δ plays in metabolism, inflammation, and cancer.
Collapse
Affiliation(s)
- Yi Liu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 426, Houston, TX 77030-4009, USA.
| | - Jennifer K Colby
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 426, Houston, TX 77030-4009, USA.
| | - Xiangsheng Zuo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 426, Houston, TX 77030-4009, USA.
| | - Jonathan Jaoude
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 426, Houston, TX 77030-4009, USA.
| | - Daoyan Wei
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Imad Shureiqi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 426, Houston, TX 77030-4009, USA.
| |
Collapse
|
288
|
Knobloch M, Widmann C. Burning fat to keep your stem cells? The role of fatty acid oxidation in various tissue stem cells. Curr Opin Lipidol 2018; 29:426-427. [PMID: 30153135 DOI: 10.1097/mol.0000000000000546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Marlen Knobloch
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | | |
Collapse
|
289
|
Santos AJM, Lo YH, Mah AT, Kuo CJ. The Intestinal Stem Cell Niche: Homeostasis and Adaptations. Trends Cell Biol 2018; 28:1062-1078. [PMID: 30195922 DOI: 10.1016/j.tcb.2018.08.001] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/02/2018] [Accepted: 08/07/2018] [Indexed: 12/20/2022]
Abstract
The intestinal epithelium is a rapidly renewing cellular compartment. This constant regeneration is a hallmark of intestinal homeostasis and requires a tightly regulated balance between intestinal stem cell (ISC) proliferation and differentiation. Since intestinal epithelial cells directly contact pathogenic environmental factors that continuously challenge their integrity, ISCs must also actively divide to facilitate regeneration and repair. Understanding niche adaptations that maintain ISC activity during homeostatic renewal and injury-induced intestinal regeneration is therefore a major and ongoing focus for stem cell biology. Here, we review recent concepts and propose an active interconversion of the ISC niche between homeostasis and injury-adaptive states that is superimposed upon an equally dynamic equilibrium between active and reserve ISC populations.
Collapse
Affiliation(s)
- António J M Santos
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yuan-Hung Lo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amanda T Mah
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
290
|
Bi S, Wang H, Kuang W. Stem cell rejuvenation and the role of autophagy in age retardation by caloric restriction: An update. Mech Ageing Dev 2018; 175:46-54. [PMID: 30031008 DOI: 10.1016/j.mad.2018.07.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 07/17/2018] [Accepted: 07/17/2018] [Indexed: 12/14/2022]
Abstract
Stem cells being pluripotent in nature can differentiate into a wide array of specific cells and asymmetrically divide to produce new ones but may undergo aging by themselves. Aging has both quantitative and qualitative effects on stem cells, and could eventually restrain them from replenishing into progenitor cells. Reactive oxygen species (ROS) accumulated in the aging cells could not only block the cell cycle but also affect autophagy by damaging the mitochondria. Autophagy could eliminate redundant production of ROS in aging stem cells and helps to maintain the proliferation capacity by restraining the expression of p16INK4a. Current studies showed that improving autophagy could restore the regenerative ability of aging stem cells. Therefore, it is important for an organism to maintain the appropriate autophagy. Caloric restriction (CR) was shown to retard the stem cell aging by a certain basic level of autophagy, suggesting that CR was an effective way to extend longevity in mammals. However, little is known about the underlying mechanisms. In this review, we tried to explore the molecular mechanisms on how CR induces appropriate autophagy to restore aging stem cell regenerative ability.
Collapse
Affiliation(s)
- Shanrong Bi
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hanyu Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weihong Kuang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
291
|
Abstract
The United States has the most expensive healthcare system worldwide. Yet measures of health span and life expectancy are well below the major industrialized nations. With the U.S. population aged 65 years and older projected to double by mid-century, a healthcare crisis is looming. Within this context, huge interest and investment have emerged in technologies and drugs to address aging with an expected benefit to health span. The thesis being that such basic interventions will reduce morbidity caused by many chronic diseases wherein biological age itself is the major risk factor. In the light of limited progress to date, a recent study out of the Harvard School of Public Health is quite refreshing: less than half dozen lifestyle interventions can greatly increase health span. Perhaps these are familiar: cessation of smoking, ≥30 minutes of moderate daily exercise, high-quality diet (limited processed food), modest alcohol intake, and maintenance of an optimal body mass index of 18.5-24.9 kg/m2. From age 50 years, women engaging in all of these behaviors versus those who do zero can expect to have a life expectancy of 43.1 additional years (an extra 14 years) with men gaining 37.6 years (an extra 12.2 years). A regimen to extend life expectancy is at hand. However, there is room for optimization by including the effects of sleep, intermittent fasting, and/or caloric restriction. Moreover, the extension of life expectancy by adherence to a healthy lifestyle revises the health span threshold for antiaging treatments under development and should provide a better set of controls for clinical trials investigating novel treatments of aging.
Collapse
Affiliation(s)
- James W Larrick
- 1 Panorama Research Institute , Sunnyvale, California.,2 Regenerative Sciences Institute , Sunnyvale, California
| | - Andrew R Mendelsohn
- 1 Panorama Research Institute , Sunnyvale, California.,2 Regenerative Sciences Institute , Sunnyvale, California
| |
Collapse
|