Abstract
Purpose of Review
Diet has profound impacts on health and longevity. Evidence is emerging to suggest that diet impinges upon the metabolic pathways in tissue-specific stem cells to influence health and disease. Here, we review the similarities and differences in the metabolism of stem cells from several tissues, and highlight the mitochondrial metabolic checkpoint in stem cell maintenance and aging. We discuss how diet engages the nutrient sensing metabolic pathways and impacts stem cell maintenance. Finally, we explore the therapeutic implications of dietary and metabolic regulation of stem cells.
Recent findings
Stem Cell transition from quiescence to proliferation is associated with a metabolic switch from glycolysis to mitochondrial OXPHOS and the mitochondrial metabolic checkpoint is critically controlled by the nutrient sensors SIRT2, SIRT3, and SIRT7 in hematopoietic stem cells. Intestine stem cell homeostasis during aging and in response to diet is critically dependent on fatty acid metabolism and ketone bodies and is influenced by the niche mediated by the nutrient sensor mTOR.
Summary
Nutrient sensing metabolic pathways critically regulate stem cell maintenance during aging and in response to diet. Elucidating the molecular mechanisms underlying dietary and metabolic regulation of stem cells provides novel insights for stem cell biology and may be targeted therapeutically to reverse stem cell aging and tissue degeneration.
Keywords: SIRT2, SIRT3, SIRT7, mTOR, calorie restriction, stem cell metabolism
Introduction
Diet exerts major effects on health and longevity. Calorie restriction (CR) is the most effective dietary intervention to extend lifespan and has been actively studied for almost a century. Pioneering research from McCay and colleagues showed that CR, a dietary regimen with a long term reduction of dietary intake for 20–40% without malnutrition, is a nutritional approach that makes rats live longer (1). Since then, other research groups have demonstrated that CR not only delays aging but ameliorates the development of age-associated diseases (2). Besides CR, intermittent fasting and methionine restriction have also been described to extend lifespan and prevent chronic diseases (3,4).
Conversely, overnutrition through a high fat diet (HFD) can accelerate aging, as it has detrimental metabolic consequences and promotes multiple aging-associated diseases such as diabetes, cancer, and cardiovascular disease. HFD, which is composed of 45% fat compared to about 10% fat in a normal diet, was first described in 1959 as a diet causing obesity due to an increase in energy intake in rats (5). Besides promoting obesity, HFD can also result in a phenotype in rodents similar to the human metabolic syndrome, characterized by insulin resistance, hyperglycemia, and hepatic steatosis (6). Importantly, the age at which dietary restriction or hypercaloric diet is started and the degree of restriction or excess define the extent of lifespan extension or reduction respectively (7).
It was thought that diet impacts health and longevity by passively changing the metabolic rate and therefore the production of the free radicals, a cause of aging and numerous diseases (8,9). However, recent advances support the view that the physiological changes triggered by diet is an actively regulated process. Prominently, the activity of nutrient sensitive metabolic regulators is modulated by diet and elicit profound physiological responses by regulating diverse metabolic pathways (10). The observations that nutrient sensitive metabolic regulators are critically required for the maintenance of tissue-specific stem cells raise an interesting possibility that nutrient sensors mediate the physiological effects of diet in part by modulating stem cell maintenance and tissue homeostasis (11–14).
In this review, we summarize the recent advances in the metabolic regulation of tissue-specific stem cells and highlight the mitochondrial metabolic checkpoint regulating stem cell maintenance. We discuss how diet affects the metabolism of stem cells through the nutrient sensing pathways, thereby influencing stem cell fate decision and tissue regeneration. Manipulation of these metabolic and nutrient sensing pathways via therapeutic interventions holds the promise for improving stem cell function and ameliorating aging-related tissue degeneration.
Metabolic Regulation in Stem Cells
The cell cycle state is a determining factor for the metabolic status of stem cells. Once animals enter the adulthood, stem cells in many tissues, such as the hematopoietic stem cells (HSCs) and neural stem cells (NSCs), mostly remain quiescent and only enter the cell cycle and proliferate periodically to self-renew and replenish the tissue (15–19). Quiescent stem cells have low mitochondrial activity and primarily rely on glycolysis for energy production (20–22). Compared to mitochondrial oxidative phosphorylation (OXPHOS), glycolysis leads to less ATP production, but is sufficient to sustain the low energy demand of quiescent stem cells. Reliance on glycolysis bypasses mitochondrial OXPHOS and protects stem cells from reactive oxygen species (ROS), and ensure long term stem cell maintenance, as elevated levels of ROS lead to loss of stem cell quiescence, differentiation, and apoptosis (11,19,23–29).
Stem cell transition from quiescence to proliferation is associated with increased mitochondrial biogenesis and a metabolic switch from glycolysis to OXPHOS to meet the increased energy demand of proliferating cells (22,30,31). This metabolic switch is critical to maintain stem cell self-renewal and function, as deletion of PTPMT1, a PTEN-like mitochondrial phosphatase, causes changes in the cell cycle and a block in differentiation, and an expansion of the HSC pool (30). The importance of the metabolic switch is also supported by the extensive studies of the PI3K/AKT/mTOR pathway, a central metabolic pathway that regulate cell growth and proliferation. Aberrant activation of the PI3K/AKT/mTOR pathway leads to loss of stem cell quiescence and the depletion of the stem cell pool, while enforced inhibition of the pathway results in persisted stem cell quiescence and impaired regenerative function (24,32–36) (Figure 1).
Figure 1. Stem Cell Metabolism and Diet.
Quiescent stem cells rely on glycolysis to prevent oxidative stress. Upon the mitogenic signals and the activation of mTOR, stem cells enter cell cycle and there is an increase in mitochondrial biogenesis and a metabolic switch from glycolysis to OXPHOS. The mitochondrial metabolic checkpoint at the restriction point monitoring the mitochondrial health is critically regulated by sirtuins (SIRT2, SIRT3, and SIRT7). The activity of sirtuins and mTOR is altered during aging, contributing to aging-associated loss of stem cell function and tissue degeneration. Diet can also change the activity of sirtuins and mTOR to regulate stem cell maintenance. Lgr5+ ISCs are critically dependent on lipid metabolism and respond to diet via PPAR. Solid lines and arrows indicate established links in stem cells. Dash lines and arrows indicate speculated links in stem cells based on studies in non-stem cells.
While stem cells from many tissues are mostly found in a quiescent state to prevent oxidative stress and maintain stem cell integrity, intestine stem cells (ISCs), which are crypt base columnar cells expressing the WNT target gene Lgr5, are highly proliferative (37). Consistent with the proliferative status, Lgr5+ ISCs have abundant mitochondria and are dependent on OXPHOS for the production of ATP and ROS, which act as signaling molecules to drive cellular differentiation (38). Lgr5+ ISCs quiescence can be induced in vitro and quiescent Lgr5+ ISCs acquire gene expression pattern indicative of reduced energy metabolism (39). A second ISC population, called +4 ISCs, is located four cells above the base of the crypt and represents a quiescent population of reserve stem cells (40). Quiescent +4 ISCs and proliferative Lgr5+ ISCs can be interconverted (41,42), suggesting that the metabolic paradigm observed in HSCs and NSCs could be conserved in ISCs. Studies on metabolic regulation in +4 ISCs and other quiescent ISC populations are much needed.
The metabolism of stem cells is influenced by the niche, the microenvironment stem cells reside in. HSCs and NSCs reside in the hypoxic niches, which keep the stem cells in a quiescent state and engage in glycolysis (43–47). Indeed, ablation of HIF1α, the master transcriptional regulator of cellular response to hypoxia, or its downstream mediator pyruvate dehydrogenase kinase (PDK), leads to loss of stem cell quiescence and exhaustion (43,48). Paneth cells, which are in direct contact with Lgr5+ ISCs, constitute a major component of the ISC niche (37). Paneth cells produce lactate through glycolysis, which is back converted to pyruvate and used to fuel the mitochondrial OXPHOS in ISCs (38).
Mitochondrial Metabolic Checkpoint Regulation of Stem Cell Maintenance
The metabolic switch from glycolysis in quiescent stem cells to mitochondrial OXPHOS in proliferative stem cells supports the concept of the mitochondrial metabolic checkpoint: because a major cellular event during the transition from quiescence to proliferation is increased mitochondrial biogenesis, a cellular condition that is monitored at the restriction point of the cell cycle is the mitochondrial health, to ensure that only cells with healthy mitochondria and the capacity to meet the energy demand of the proliferative cells can enter the cell cycle (49). Consistent with this model, impaired mitochondrial function due to LKB1 deletion results in loss of HSC quiescence, HSC death, compromised HSC repopulation capacity, and defective hematopoiesis (50–52), and perturbation of mitochondrial function by ablation of the mitochondrial transcription factor A (TFAM) leads to neurogenesis defects (53).
Recent studies reveal that the mitochondrial metabolic checkpoint is critically controlled by sirtuins, a family of NAD+-dependent deacetylases (Figure 1). SIRT3, a mitochondrial deacetylase, monitors mitochondrial oxidative stress and improves oxidative stress resistance, while SIRT7, a histone deacetylase, monitors mitochondrial protein folding stress and regulates mitochondrial unfolded protein response (11,12,54–56). Consistent with a role in monitoring the mitochondrial metabolic checkpoint at the restriction point, deletion of SIRT3 or SIRT7 results in loss of HSC quiescence, increased cell death, and compromised HSC function (11,12). Mitochondrial stress triggers HSC death that is mediated by the NLRP3 inflammasome (13). SIRT2 suppresses the activation of the NLRP3 inflammasome and prevents mitochondrial stress-induced HSC death (13,57). Mitochondrial unfolded protein response is also a conserved regulatory mechanism of stem cell maintenance across tissues (12,58,59). Other mitochondrial protective mechanisms in stem cells include mitophagy (60) and mitochondrial fusion and fission (61,62).
Dietary Regulation of Stem Cells
Given its location, the intestinal epithelium constantly encounters diet-derived nutrients and is strongly responsive to changes in nutrient intake (63). CR leads to reduced small intestine mass, shorter villi, and fewer enterocytes (64). While the number and proliferation of Lgr5+ ISCs are increased, proliferation of transit amplifying cells, more differentiated progenitors, is reduced in CR mice. Thus, CR supports self-renewal of Lgr5+ ISCs at the expense of differentiation. Short term fasting does not change the number of Lgr5+ ISCs but enhances their function (65). Surprisingly, HFD is also associated with shorter small intestine with reduced intestine weight, and similar morphologic modifications of the intestinal epithelium induced by CR, including a reduction in villi length, a decrease in the number of enterocytes, and an increase in crypt depth (66). HFD causes an increase in both the number and function of ISCs. Despite at the opposite spectrum of calorie intake, CR/fasting and HFD are both associated with increased fatty acid metabolism. The similar effects of these dietary regimens on ISC maintenance and intestine structure may reflect a central role of fatty acid metabolism in ISC homeostasis.
Indeed, the effect of HFD on ISCs is mediated by certain fatty acids contained in HFD, which activate a PPAR-δ transcriptional program, leading to the activation of the Wnt/β-catenin pathway, a central regulator of ISC self-renewal (66). Similarly, short term fasting increases ISC and progenitor cell function through the induction of the fatty acid oxidation program downstream of PPAR signaling (65). 3-hydroxy-3-methylglutaryl-coenzyme A [CoA] synthetase 2 (Hmgcs2), which encodes the rate-limiting enzyme in the production of ketone bodies, is highly expressed in ISCs. The production of ketone bodies modulates the Notch signaling, which promotes ISC self-renewal and instructs lineage differentiation (67). The Hmgcs2 signaling in ISCs is enhanced by CR, contributing to improved ISC self-renewal and reduced differentiation (68).
In addition to directly impacting the metabolic pathways in ISCs, diet can also modulate ISC homeostasis through the niche. The effects observed on increased ISC number and proliferation during CR are associated with a modulation of the mTOR signaling in Paneth cells (64). CR reduces mTOR activity in Paneth cells, leading to the induction of the ectoenzyme Bst1 and secretion of the paracrine factor cyclic ADP ribose (cADPR), which promotes ISC self-renewal and expansion. Contrary to CR, under HFD, ISCs can grow in the absence of signals from Paneth cells through the activation of the Wnt signaling, allowing them to adjust to the decrease number of Paneth cells (66) (Figure 1).
Diet also impacts the HSC and NSC compartments, although the molecular mechanisms are largely unknown. Unlike ISCs, CR/fasting and HFD have opposite effects on HSCs and NSCs. CR increases HSC quiescence and improves HSC function (69,70). As to NSCs, CR enhances neurogenesis and memory performance (71–75). Prolonged fasting or intermittent fasting also give similar beneficial effects on HSCs (76,77) and NSCs (78–80). Conversely, HFD causes loss of HSC quiescence, loss of the HSC pool, promotes the expansion of myeloid progenitors, renders increased susceptibility to myeloablative stress and reduced HSC reconstitute capacity (81–84), as well as reduced neurogenesis (85,86). Much work is needed to understand how diet regulates metabolism in HSCs and NSCs, and whether the mitochondrial metabolic checkpoint is affected.
Therapeutic potential
The regenerative potential of stem cells declines during aging, accounting for much of aging-associated tissue degeneration and dysfunction (87–89). Recent studies attribute stem cell aging to dysregulated nutrient sensing metabolic pathway (11–13,90), which act as the mediators of diet-induced physiological changes (55,57,64,66,91,92), suggesting that the nutrient sensing metabolic pathways can be targeted therapeutically to mimic the physiological effects of diet on preventing stem cell aging and tissue degeneration (Figure 1).
Mitochondrial oxidative stress and protein folding stress are increased in aged HSCs (12,28), and increased mitochondrial stress results in loss of HSC quiescence, increased cell death, reduced repopulation capacity, and myeloid-biased differentiation, resembling essential aspects of HSC aging (11,12,25,27). The expression of SIRT2, SIRT3, and SIRT7 is reduced in aged HSCs, resulting in increased mitochondrial stress and NLRP3-mediated cell death (11–13,93). Thus, HSC aging results from the dysregulation of the mitochondrial metabolic checkpoint. The activity of mTOR is increased in aged Paneth cells, which inhibits the activity of PPAR-α and the production of Notum, an extracellular inhibitor of the stemness-maintaining Wnt signaling (90). Importantly, genetic manipulations of these nutrient sensors or downstream mediators reverse stem cell aging, highlighting the reversibility and therapeutic potential of aging-associated stem cell decline and tissue degeneration (11–13,90).
Evidence for pharmacological manipulation of these pathways is emerging. The activity of sirtuins is critically dependent on NAD+ and the supplementation of nicotinamide riboside, a precursor of NAD+, reduces mitochondrial stress and improves stem cell maintenance across tissues (58,94,95). Rapamycin, an inhibitor of mTOR, also promotes the function of many tissue-specific stem cells (33,64). An inhibitor of Notum enhances the function of aged ISCs and promotes the recovery from injury (90). These observations support the notion that these nutrient sensing metabolic pathways can be targeted therapeutically as CR mimetics, which bring the benefits of CR without a dietary regimen that cannot be tolerated by most individuals.
Conclusions
The burgeoning research area of metabolic regulation in stem cell homeostasis is underscored by its physiological impacts and therapeutic potentials. Recent studies have revealed shared and unique metabolic regulatory mechanisms in stem cells from various tissues. A metabolic switch from glycolysis in quiescent stem cells to OXPHOS in proliferative stem cells appears to be a shared mechanism in stem cells of many tissues. The mitochondrial metabolic checkpoint at the restriction point is critical to ensure that only stem cells with healthy mitochondria and the capacity to meet the energy demand of the proliferative cells can enter the cell cycle. Highly proliferative ISCs are critically dependent on fatty acid metabolism and ketone bodies. In addition to cell-intrinsic metabolic regulatory mechanisms, stem cell metabolism is also influenced by the niche.
Stem cell metabolism regulated by diet is just emerging, opening the door for novel insights into stem cell biology. Nutrient sensing metabolic pathways critically regulate stem cell maintenance during aging and in response to diet. Elucidating the molecular mechanisms underlying dietary and metabolic regulation of stem cells provides novel insights for stem cell biology and may be targeted therapeutically to reverse stem cell aging and tissue degeneration.
Acknowledgements
Supported by NIH R01DK 117481 (D.C.), R01DK101885 (D.C.), R01AG063404 (D.C.), R01AG 063389 (D.C.), National Institute of Food and Agriculture (D.C.).
Footnotes
Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of a an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version.
Conflict of Interest Marine Barthez, Zehan Song, Chih Ling Wang, and Danica Chen have no conflict of interest.
Contributor Information
Marine Barthez, Program in Metabolic Biology, Nutritional Sciences & Toxicology, University of California, Berkeley, CA 94720.
Zehan Song, Program in Metabolic Biology, Nutritional Sciences & Toxicology, University of California, Berkeley, CA 94720.
Chih Ling Wang, Program in Metabolic Biology, Nutritional Sciences & Toxicology, University of California, Berkeley, CA 94720.
Danica Chen, Program in Metabolic Biology, Nutritional Sciences & Toxicology, University of California, Berkeley, CA 94720, 119 Morgan Hall, University of California, Berkeley, CA 94720.
References
Papers of particular interest, published recently, have been highlighted as:
* Of importance
- 1.McCay CM, Crowell MF, Maynard LA. The Effect of Retarded Growth Upon the Length of Life Span and Upon the Ultimate Body Size. [Internet]. The Journal of Nutrition 1935. [cited 2020 May 19]. p. 63–79. [PubMed] [Google Scholar]
- 2.Weindruch R and Walford RL. The retardation of aging and disease by dietary restriction. CC Thomas, Springfield, IL. 1988. [Google Scholar]
- 3.Carlson AJ, Hoelzel F. Apparent Prolongation of the Life Span of Rats by Intermittent Fasting - PubMed [Internet]. J Nutr. . 1946. [cited 2020 May 19]. p. 363–75. [DOI] [PubMed] [Google Scholar]
- 4.Orentreich N, Matias JR, DeFelice A, Zimmerman JA. Low Methionine Ingestion by Rats Extends Life Span - PubMed [Internet]. J Nutr. 1993. [cited 2020 May 19]. p. 269–74. [DOI] [PubMed] [Google Scholar]
- 5.Mašek J, Fábry P. High-fat diet and the development of obesity in albino rats. Experientia. 1959. November;15(11):444–5. [DOI] [PubMed] [Google Scholar]
- 6.Wang CY, Liao JK. A mouse model of diet-induced obesity and insulin resistance. Methods Mol Biol. 2012;821:421–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Fontana L, Partridge L, Longo VD. Extending healthy life span-from yeast to humans. Vol. 328, Science. Science; 2010. p. 321–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sohal RS, Weindruch R. Oxidative stress, caloric restriction, and aging. Science (80-). 1996; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.HARMAN D Aging: a theory based on free radical and radiation chemistry. J Gerontol. 1956; [DOI] [PubMed] [Google Scholar]
- 10.Luo H, Chiang HH, Louw M, Susanto A, Chen D. Nutrient Sensing and the Oxidative Stress Response. Trends in Endocrinology and Metabolism. 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Brown K, Xie S, Qiu X, Mohrin M, Shin J, Liu Y, et al. SIRT3 Reverses Aging-Associated Degeneration. Cell Rep. 2013. February;3(2):319–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mohrin M, Shin J, Liu Y, Brown K, Luo H, Xi Y, et al. A mitochondrial UPR-mediated metabolic checkpoint regulates hematopoietic stem cell aging. Science (80-). 2015. March;347(6228):1374–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. *.Luo H, Mu WC, Karki R, Chiang HH, Mohrin M, Shin JJ, et al. Mitochondrial Stress-Initiated Aberrant Activation of the NLRP3 Inflammasome Regulates the Functional Deterioration of Hematopoietic Stem Cell Aging. Cell Rep. 2019;26(4):945–954.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]; Demonstrates that mitochondrial stress leads to stem cell aging through the NRLP3 inflammasome activation and caspase 1-mediated cell death.
- 14.Chen C, Liu Y, Liu R, Ikenoue T, Guan KL, Liu Y, et al. TSC-mTOR maintains quiescence and function of hematopoietic stem cells by repressing mitochondrial biogenesis and reactive oxygen species. J Exp Med. 2008; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Cheshier SH, Morrison SJ, Liao X, Weissman IL. In vivo proliferation and cell cycle kinetics of long-term self-renewing hematopoietic stem cells. Proc Natl Acad Sci U S A. 1999; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lucassen PJ, Meerlo P, Naylor AS, van Dam AM, Dayer AG, Fuchs E, et al. Regulation of adult neurogenesis by stress, sleep disruption, exercise and inflammation: Implications for depression and antidepressant action. Eur Neuropsychopharmacol [Internet]. 2010;20(1):1–17. [DOI] [PubMed] [Google Scholar]
- 17.Wang YAZ, Plane JM, Jiang P, Zhou CJ, Deng W. Concise review: Quiescent and active states of endogenous adult neural stem cells: Identification and characterization. Stem Cells. 2011;29(6):907–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ding WY, Huang J, Wang H. Waking up quiescent neural stem cells: Molecular mechanisms and implications in neurodevelopmental disorders. PLOS Genet [Internet]. 2020. April;16(4):e1008653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Cavallucci V, Fidaleo M, Pani G. Neural Stem Cells and Nutrients: Poised Between Quiescence and Exhaustion. Vol. 27, Trends in Endocrinology and Metabolism. Elsevier Inc.; 2016. p. 756–69. [DOI] [PubMed] [Google Scholar]
- 20.Simsek T, Kocabas F, Zheng J, Deberardinis RJ, Mahmoud AI, Olson EN, et al. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell. 2010; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ito K, Suda T. Metabolic requirements for the maintenance of self-renewing stem cells. Vol. 15, Nature Reviews Molecular Cell Biology. Nature Publishing Group; 2014. p. 243–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Shin J, Berg DA, Zhu Y, Shin JY, Song J, Bonaguidi MA, et al. Single-Cell RNA-Seq with Waterfall Reveals Molecular Cascades underlying Adult Neurogenesis. Cell Stem Cell [Internet]. 2015;17(3):360–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Maryanovich M, Zaltsman Y, Ruggiero A, Goldman A, Shachnai L, Zaidman SL, et al. An MTCH2 pathway repressing mitochondria metabolism regulates haematopoietic stem cell fate. Nat Commun. 2015. July 29;6. [DOI] [PubMed] [Google Scholar]
- 24.Juntilla MM, Patil VD, Calamito M, Joshi RP, Birnbaum MJ, Koretzky GA. AKT1 and AKT2 maintain hematopoietic stem cell function by regulating reactive oxygen species. Blood. 2010;115(20):4030–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Tothova Z, Kollipara R, Huntly BJ, Lee BH, Castrillon DH, Cullen DE, et al. FoxOs Are Critical Mediators of Hematopoietic Stem Cell Resistance to Physiologic Oxidative Stress. Cell. 2007;128(2):325–39. [DOI] [PubMed] [Google Scholar]
- 26.Owusu-Ansah E, Banerjee U. Reactive oxygen species prime Drosophila haematopoietic progenitors for differentiation. Nature. 2009; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ito K, Hirao A, Arai F, Matsuoka S, Takubo K, Hamaguchi I, et al. Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells. Nature. 2004;431(7011):997–1002. [DOI] [PubMed] [Google Scholar]
- 28.Ito K, Hirao A, Arai F, Takubo K, Matsuoka S, Miyamoto K, et al. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat Med. 2006;12(4):446–51. [DOI] [PubMed] [Google Scholar]
- 29.Miyamoto K, Araki KY, Naka K, Arai F, Takubo K, Yamazaki S, et al. Foxo3a Is Essential for Maintenance of the Hematopoietic Stem Cell Pool. Cell Stem Cell. 2007; [DOI] [PubMed] [Google Scholar]
- 30.Yu WM, Liu X, Shen J, Jovanovic O, Pohl EE, Gerson SL, et al. Metabolic regulation by the mitochondrial phosphatase PTPMT1 is required for hematopoietic stem cell differentiation. Cell Stem Cell. 2013. January 3;12(1):62–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Mohrin M, Widjaja A, Liu Y, Luo H, Chen D. The mitochondrial unfolded protein response is activated upon hematopoietic stem cell exit from quiescence. Aging Cell. 2018;17(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Haneline LS, White H, Yang FC, Chen S, Orschell C, Kapur R, et al. Genetic reduction of class Ia PI-3 kinase activity alters fetal hematopoiesis and competitive repopulating ability of hematopoietic stem cells in vivo. Blood. 2006; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Chen C, Liu Y, Liu Y, Zheng P. MTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells. Sci Signal. 2009;2(98). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Magri L, Cambiaghi M, Cominelli M, Alfaro-Cervello C, Cursi M, Pala M, et al. Sustained activation of mTOR pathway in embryonic neural stem cells leads to development of tuberous sclerosis complex-associated lesions. Cell Stem Cell. 2011; [DOI] [PubMed] [Google Scholar]
- 35.Kassai H, Sugaya Y, Noda S, Nakao K, Maeda T, Kano M, et al. Selective Activation of mTORC1 Signaling Recapitulates Microcephaly, Tuberous Sclerosis, and Neurodegenerative Diseases. Cell Rep. 2014; [DOI] [PubMed] [Google Scholar]
- 36.Paliouras GN, Hamilton LK, Aumont A, Joppé SE, Barnabé-Heider F, Fernandes KJL. Mammalian target of rapamycin signaling is a key regulator of the transit-amplifying progenitor pool in the adult and aging forebrain. J Neurosci. 2012; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Barker N, Van Es JH, Kuipers J, Kujala P, Van Den Born M, Cozijnsen M, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007. October 25;449(7165):1003–7. [DOI] [PubMed] [Google Scholar]
- 38. *.Rodríguez-Colman MJ, Schewe M, Meerlo M, Stigter E, Gerrits J, Pras-Raves M, et al. Interplay between metabolic identities in the intestinal crypt supports stem cell function. Nature. 2017. March 16;543(7645):424–7. [DOI] [PubMed] [Google Scholar]; Shows highly proliferative intestine stem cells rely on mitochondrial OXPHOS for ATP production, which is supported by glycolysis in the niche.
- 39.Basak O, Beumer J, Wiebrands K, Seno H, van Oudenaarden A, Clevers H. Induced Quiescence of Lgr5+ Stem Cells in Intestinal Organoids Enables Differentiation of Hormone-Producing Enteroendocrine Cells. Cell Stem Cell. 2017; [DOI] [PubMed] [Google Scholar]
- 40.Li N, Clevers H. Coexistence of quiescent and active adult stem cells in mammals. Vol. 327, Science. Science; 2010. p. 542–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Takeda N, Jain R, LeBoeuf MR, Wang Q, Lu MM, Epstein JA. Interconversion between intestinal stem cell populations in distinct niches. Science (80-). 2011; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Tian H, Biehs B, Warming S, Leong KG, Rangell L, Klein OD, et al. A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable. Nature. 2011; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Takubo K, Goda N, Yamada W, Iriuchishima H, Ikeda E, Kubota Y, et al. Regulation of the HIF-1α level is essential for hematopoietic stem cells. Cell Stem Cell. 2010;7(3):391–402. [DOI] [PubMed] [Google Scholar]
- 44.Takubo K, Nagamatsu G, Kobayashi CI, Nakamura-Ishizu A, Kobayashi H, Ikeda E, et al. Regulation of glycolysis by Pdk functions as a metabolic checkpoint for cell cycle quiescence in hematopoietic stem cells. Cell Stem Cell. 2013;12(1):49–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Pistollato F, Chen H-L, Schwartz PH, Basso G, Panchision DM. Oxygen tension controls the expansion of human CNS precursors and the generation of astrocytes and oligodendrocytes. Mol Cell Neurosci [Internet]. 2007. July;35(3):424–35. [DOI] [PubMed] [Google Scholar]
- 46.Moreno M, Fernández V, Monllau JM, Borrell V, Lerin C, de la Iglesia N. Transcriptional Profiling of Hypoxic Neural Stem Cells Identifies Calcineurin-NFATc4 Signaling as a Major Regulator of Neural Stem Cell Biology. Stem cell reports [Internet]. 2015/07/30. 2015. August;5(2):157–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kim DY, Rhee I, Paik J. Metabolic circuits in neural stem cells. Cell Mol Life Sci [Internet]. 2014/07/19. 2014. November;71(21):4221–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Takubo K, Nagamatsu G, Kobayashi CI, Nakamura-Ishizu A, Kobayashi H, Ikeda E, et al. Regulation of glycolysis by Pdk functions as a metabolic checkpoint for cell cycle quiescence in hematopoietic stem cells. Cell Stem Cell. 2013. January 3;12(1):49–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Mohrin M, Chen D. The mitochondrial metabolic checkpoint and aging of hematopoietic stem cells. Current Opinion in Hematology. 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Gurumurthy S, Xie SZ, Alagesan B, Kim J, Yusuf RZ, Saez B, et al. The Lkb1 metabolic sensor maintains haematopoietic stem cell survival. Nature. 2010; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gan B, Hu J, Jiang S, Liu Y, Sahin E, Zhuang L, et al. Lkb1 regulates quiescence and metabolic homeostasis of haematopoietic stem cells. Nature. 2010; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Nakada D, Saunders TL, Morrison SJ. Lkb1 regulates cell cycle and energy metabolism in haematopoietic stem cells. Nature. 2010; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Beckervordersandforth R, Ebert B, Schäffner I, Moss J, Fiebig C, Shin J, et al. Role of Mitochondrial Metabolism in the Control of Early Lineage Progression and Aging Phenotypes in Adult Hippocampal Neurogenesis. Neuron. 2017; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Yan W, Liang Y, Zhang Q, Wang D, Lei M, Qu J, et al. Arginine methylation of SIRT 7 couples glucose sensing with mitochondria biogenesis. EMBO Rep. 2018;19(12):1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Qiu X, Brown K, Hirschey MD, Verdin E, Chen D. Calorie Restriction Reduces Oxidative Stress by SIRT3-Mediated SOD2 Activation. Cell Metab. 2010. December;12(6):662–7. [DOI] [PubMed] [Google Scholar]
- 56.Someya S, Yu W, Hallows WC, Xu J, Vann JM, Leeuwenburgh C, et al. Sirt3 mediates reduction of oxidative damage and prevention of age-related hearing loss under Caloric Restriction. Cell. 2010;143(5):802–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.He M, Chiang HH, Luo H, Zheng Z, Qiao Q, Wang L, et al. An Acetylation Switch of the NLRP3 Inflammasome Regulates Aging-Associated Chronic Inflammation and Insulin Resistance. Cell Metab. 2020;31(3):580–591.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zhang H, Ryu D, Wu Y, Gariani K, Wang X, Luan P, et al. NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science (80-). 2016. June 17;352(6292):1436–43. [DOI] [PubMed] [Google Scholar]
- 59.Berger E, Rath E, Yuan D, Waldschmitt N, Khaloian S, Allgäuer M, et al. Mitochondrial function controls intestinal epithelial stemness and proliferation. Nat Commun. 2016; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ito K, Turcotte R, Cui J, Zimmerman SE, Pinho S, Mizoguchi T, et al. Self-renewal of a purified Tie2+ hematopoietic stem cell population relies on mitochondrial clearance. Science (80-). 2016; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Khacho M, Clark A, Svoboda DS, Azzi J, MacLaurin JG, Meghaizel C, et al. Mitochondrial Dynamics Impacts Stem Cell Identity and Fate Decisions by Regulating a Nuclear Transcriptional Program. Cell Stem Cell. 2016; [DOI] [PubMed] [Google Scholar]
- 62.Luchsinger LL, De Almeida MJ, Corrigan DJ, Mumau M, Snoeck HW. Mitofusin 2 maintains haematopoietic stem cells with extensive lymphoid potential. Nature. 2016; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Moog F The lining of the small intestine. Sci Am. 1981;245(5):154–76. [DOI] [PubMed] [Google Scholar]
- 64.Yilmaz ÖH, Katajisto P, Lamming DW, Gültekin Y, Bauer-Rowe KE, Sengupta S, et al. MTORC1 in the Paneth cell niche couples intestinal stem-cell function to calorie intake. Nature. 2012. June 28;486(7404):490–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. *.Mihaylova MM, Cheng CW, Cao AQ, Tripathi S, Mana MD, Bauer-Rowe KE, et al. Fasting Activates Fatty Acid Oxidation to Enhance Intestinal Stem Cell Function during Homeostasis and Aging. Cell Stem Cell. 2018. May 3;22(5):769–778.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]; Demonstrates intestine stem cells critically depends on fatty acid metabolism and is responsive to diet.
- 66. *.Beyaz S, Mana MD, Roper J, Kedrin D, Saadatpour A, Hong SJ, et al. High-fat diet enhances stemness and tumorigenicity of intestinal progenitors. Nature. 2016. March 2;531(7592):53–8. [DOI] [PMC free article] [PubMed] [Google Scholar]; Demonstrates intestine stem cells critically depends on fatty acid metabolism and is responsive to diet.
- 67. *.Cheng CW, Biton M, Haber AL, Gunduz N, Eng G, Gaynor LT, et al. Ketone Body Signaling Mediates Intestinal Stem Cell Homeostasis and Adaptation to Diet. Cell. 2019. August 22;178(5):1115–1131.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]; Demonstrates intestine stem cells critically depends on ketone bodies and is responsive to diet.
- 68.Gebert N, Cheng CW, Kirkpatrick JM, Di Fraia D, Yun J, Schädel P, et al. Region-Specific Proteome Changes of the Intestinal Epithelium during Aging and Dietary Restriction. Cell Rep. 2020. April 28;31(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Chen J, Astle CM, Harrison DE. Hematopoietic senescence is postponed and hematopoietic stem cell function is enhanced by dietary restriction. Exp Hematol. 2003;31(11):1097–103. [DOI] [PubMed] [Google Scholar]
- 70.Tang D, Tao S, Chen Z, Koliesnik IO, Calmes PG, Hoerr V, et al. Dietary restriction improves repopulation but impairs lymphoid differentiation capacity of hematopoietic stem cells in early aging. J Exp Med. 2016;213(4):535–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Apple DM, Mahesula S, Fonseca RS, Zhu C, Kokovay E. Calorie restriction protects neural stem cells from age-related deficits in the subventricular zone. Aging (Albany NY) [Internet]. 2019. January;11(1):115–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lee J, Duan W, Long JM, Ingram DK, Mattson MP. Dietary restriction increases the number of newly generated neural cells, and BDNF expression, in the dentate gyrus of rats. J Mol Neurosci. 2000;15(2):99–108. [DOI] [PubMed] [Google Scholar]
- 73.Kaptan Z, Akgün-Dar K, Kapucu A, Dedeakayoğullari H, Batu Ş, Üzüm G. Long term consequences on spatial learning-memory of low-calorie diet during adolescence in female rats; hippocampal and prefrontal cortex BDNF level, expression of NeuN and cell proliferation in dentate gyrus. Brain Res [Internet]. 2015;1618:194–204. [DOI] [PubMed] [Google Scholar]
- 74.Hornsby AKE, Redhead YT, Rees DJ, Ratcliff MSG, Reichenbach A, Wells T, et al. Short-term calorie restriction enhances adult hippocampal neurogenesis and remote fear memory in a Ghsr-dependent manner. Psychoneuroendocrinology [Internet]. 2015/09/25. 2016. January;63:198–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kim Y, Sehee K, Chanyang K, Takahiro S, Masayasu K, Seungjoon P. Ghrelin is required for dietary restriction-induced enhancement of hippocampal neurogenesis: lessons from ghrelin knockout mice. Endocr J [Internet]. 2015;62(3):269–75. [DOI] [PubMed] [Google Scholar]
- 76.Cheng CW, Adams GB, Perin L, Wei M, Zhou X, Lam BS, et al. Prolonged fasting reduces IGF-1/PKA to promote hematopoietic-stem-cell- based regeneration and reverse immunosuppression. Cell Stem Cell. 2014;14(6):810–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Mendelsohn AR, Larrick JW. Prolonged fasting/refeeding promotes hematopoietic stem cell regeneration and rejuvenation. Rejuvenation Res. 2014;17(4):385–9. [DOI] [PubMed] [Google Scholar]
- 78.Baik SH, Rajeev V, Fann DYW, Jo DG, Arumugam TV. Intermittent fasting increases adult hippocampal neurogenesis. Brain Behav. 2020;10(1):1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Mattson MP, Longo VD, Harvie M. Impact of intermittent fasting on health and disease processes. Ageing Res Rev [Internet]. 2016/10/31. 2017. October;39:46–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Brandhorst S, Choi IY, Wei M, Cheng CW, Sedrakyan S, Navarrete G, et al. A Periodic Diet that Mimics Fasting Promotes Multi-System Regeneration, Enhanced Cognitive Performance, and Healthspan. Cell Metab [Internet]. 2015/06/18. 2015. July;22(1):86–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Singer K, DelProposto J, Lee Morris D, Zamarron B, Mergian T, Maley N, et al. Diet-induced obesity promotes myelopoiesis in hematopoietic stem cells. Mol Metab. 2014;3(6):664–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Li Y, Zhu S, Zhang Y, Liu T, Su L, Zhang Q, et al. High fat diet-induced obesity exacerbates hematopoiesis deficiency and cytopenia caused by 5-fluorouracil via peroxisome proliferator-activated receptor γ. Exp Hematol. 2018;60(2018):30–39.e1. [DOI] [PubMed] [Google Scholar]
- 83.Van Den Berg SM, PSeijkens TT, HKusters PJ, Beckers L, DenToom M, Smeets E, et al. Diet-induced obesity in mice diminishes hematopoietic stem and progenitor cells in the bone marrow. FASEB J. 2016;30(5):1779–88. [DOI] [PubMed] [Google Scholar]
- 84.Hermetet F, Buffière A, Aznague A, Pais de Barros JP, Bastie JN, Delva L, et al. High-fat diet disturbs lipid raft/TGF-β signaling-mediated maintenance of hematopoietic stem cells in mouse bone marrow. Nat Commun. 2019;10(1):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Park HR, Park M, Choi J, Park K-Y, Chung HY, Lee J. A high-fat diet impairs neurogenesis: Involvement of lipid peroxidation and brain-derived neurotrophic factor. Neurosci Lett [Internet]. 2010;482(3):235–9. [DOI] [PubMed] [Google Scholar]
- 86.Robison LS, Albert NM, Camargo LA, Anderson BM, Salinero AE, Riccio DA, et al. High-fat diet-induced obesity causes sex-specific deficits in adult hippocampal neurogenesis in mice. eNeuro. 2020;7(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Vol. 153, Cell. Cell Press; 2013. p. 1194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Geiger H, De Haan G, Carolina Florian M. The ageing haematopoietic stem cell compartment. Nature Reviews Immunology. 2013. [DOI] [PubMed] [Google Scholar]
- 89.Oh J, Lee YD, Wagers AJ. Stem cell aging: Mechanisms, regulators and therapeutic opportunities. Nature Medicine. 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. *.Pentinmikko N, Iqbal S, Mana M, Andersson S, Cognetta AB, Suciu RM, et al. mNotum produced by Paneth cells attenuates regeneration of aged intestinal epitheliu. Nature. 2019. July 18;571(7765):398–402. [DOI] [PMC free article] [PubMed] [Google Scholar]; Shows intestine stem cell aging is regulated by mTOR signaling in the niche.
- 91.Chen D, Steele AD, Lindquist S, Guarente L. Medicine: Increase in activity during calorie restriction requires Sirt1. Science (80-). 2005; [DOI] [PubMed] [Google Scholar]
- 92.Shin J, He M, Liu Y, Paredes S, Villanova L, Brown K, et al. SIRT7 represses myc activity to suppress er stress and prevent fatty liver disease. Cell Rep. 2013; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Chambers SM, Shaw CA, Gatza C, Fisk CJ, Donehower LA, Goodell MA. Aging hematopoietic stem cells decline in function and exhibit epigenetic dysregulation. PLoS Biol. 2007;5(8):1750–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. *.Vannini N, Campos V, Girotra M, Trachsel V, Rojas-Sutterlin S, Tratwal J, et al. The NAD-Booster Nicotinamide Riboside Potently Stimulates Hematopoiesis through Increased Mitochondrial Clearance. Cell Stem Cell. 2019; [DOI] [PubMed] [Google Scholar]; Shows the therapeutic potential of the mitochondrial metabolic checkpoint in stem cells.
- 95.Igarashi M, Miura M, Williams E, Jaksch F, Kadowaki T, Yamauchi T, et al. NAD+ supplementation rejuvenates aged gut adult stem cells. Aging Cell. 2019. June 1; 18(3). [DOI] [PMC free article] [PubMed] [Google Scholar]