1
|
Cain TL, Derecka M, McKinney-Freeman S. The role of the haematopoietic stem cell niche in development and ageing. Nat Rev Mol Cell Biol 2025; 26:32-50. [PMID: 39256623 DOI: 10.1038/s41580-024-00770-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 09/12/2024]
Abstract
Blood production depends on rare haematopoietic stem cells (HSCs) and haematopoietic stem and progenitor cells (HSPCs) that ultimately take up residence in the bone marrow during development. HSPCs and HSCs are subject to extrinsic regulation by the bone marrow microenvironment, or niche. Studying the interactions between HSCs and their niche is critical for improving ex vivo culturing conditions and genetic manipulation of HSCs, which is pivotal for improving autologous HSC therapies and transplantations. Additionally, understanding how the complex molecular network in the bone marrow is altered during ageing is paramount for developing novel therapeutics for ageing-related haematopoietic disorders. HSCs are unique amongst stem and progenitor cell pools in that they engage with multiple physically distinct niches during their ontogeny. HSCs are specified from haemogenic endothelium in the aorta, migrate to the fetal liver and, ultimately, colonize their final niche in the bone marrow. Recent studies employing single-cell transcriptomics and microscopy have identified novel cellular interactions that govern HSC specification and engagement with their niches throughout ontogeny. New lineage-tracing models and microscopy tools have raised questions about the numbers of HSCs specified, as well as the functional consequences of HSCs interacting with each developmental niche. Advances have also been made in understanding how these niches are modified and perturbed during ageing, and the role of these altered interactions in haematopoietic diseases. In this Review, we discuss these new findings and highlight the questions that remain to be explored.
Collapse
Affiliation(s)
- Terri L Cain
- Department of Haematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Marta Derecka
- Department of Haematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|
2
|
Furuhashi K, Kakiuchi M, Ueda R, Oda H, Ummarino S, Ebralidze AK, Bassal MA, Meng C, Sato T, Lyu J, Han MG, Maruyama S, Watanabe Y, Sawa Y, Kato D, Wake H, Reizis B, Frangos JA, Owens DM, Tenen DG, Ghiran IC, Robson SC, Fujisaki J. Bone marrow niches orchestrate stem-cell hierarchy and immune tolerance. Nature 2025:10.1038/s41586-024-08352-6. [PMID: 39743593 DOI: 10.1038/s41586-024-08352-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 11/06/2024] [Indexed: 01/04/2025]
Abstract
Stem cells reside in specialized microenvironments, termed niches, at several different locations in tissues1-3. The differential functions of heterogeneous stem cells and niches are important given the increasing clinical applications of stem-cell transplantation and immunotherapy. Whether hierarchical structures among stem cells at distinct niches exist and further control aspects of immune tolerance is unknown. Here we describe previously unknown new hierarchical arrangements in haematopoietic stem cells (HSCs) and bone marrow niches that dictate both regenerative potential and immune privilege. High-level nitric oxide-generating (NOhi) HSCs are refractory to immune attack and exhibit delayed albeit robust long-term reconstitution. Such highly immune-privileged, primitive NOhi HSCs co-localize with distinctive capillaries characterized by primary ciliated endothelium and high levels of the immune-checkpoint molecule CD200. These capillaries regulate the regenerative functions of NOhi HSCs through the ciliary protein IFT20 together with CD200, endothelial nitric oxide synthase and autophagy signals, which further mediate immunoprotection. Notably, previously described niche constituents, sinusoidal cells and type-H vessels2-10 co-localize with less immune-privileged and less potent NOlow HSCs. Together, we identify highly immune-privileged, late-rising primitive HSCs and characterize their immunoprotective niches comprising specialized vascular domains. Our results indicate that the niche orchestrates hierarchy in stem cells and immune tolerance, and highlight future immunotherapeutic targets.
Collapse
Affiliation(s)
- Kazuhiro Furuhashi
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Columbia Stem Cell Initiatives, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Nagoya University Institute for Advanced Research, Nagoya, Japan
| | - Miwako Kakiuchi
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Columbia Stem Cell Initiatives, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Ryosuke Ueda
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Columbia Stem Cell Initiatives, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Center for Inflammation Research, Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hiroko Oda
- Center for Inflammation Research, Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Simone Ummarino
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Hematology and Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Alexander K Ebralidze
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Hematology and Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Mahmoud A Bassal
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
- Division of Hematology and Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Chen Meng
- Center for Inflammation Research, Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Tatsuyuki Sato
- Center for Inflammation Research, Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jing Lyu
- Center for Inflammation Research, Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Min-Guk Han
- Center for Inflammation Research, Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yu Watanabe
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuriko Sawa
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Kato
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Boris Reizis
- Translational Immunology Center, Department of Pathology, New York University, New York, NY, USA
| | | | - David M Owens
- Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Daniel G Tenen
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Hematology and Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Ionita C Ghiran
- Center for Inflammation Research, Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Simon C Robson
- Center for Inflammation Research, Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Joji Fujisaki
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
- Columbia Stem Cell Initiatives, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
- Center for Inflammation Research, Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Division of Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Becker IC, Barrachina MN, Lykins J, Camacho V, Stone AP, Chua BA, Signer RAJ, Machlus KR, Whiteheart SW, Roweth HG, Italiano JE. Inhibition of RhoA-mediated secretory autophagy in megakaryocytes mitigates myelofibrosis in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626665. [PMID: 39677616 PMCID: PMC11642871 DOI: 10.1101/2024.12.04.626665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Megakaryocytes (MKs) are large, polyploid cells that contribute to bone marrow homeostasis through the secretion of cytokines such as transforming growth factor β1 (TGFβ1). During neoplastic transformation, immature MKs accumulate in the bone marrow where they induce fibrotic remodeling ultimately resulting in myelofibrosis. Current treatment strategies aim to prevent MK hyperproliferation, however, little is understood about the potential of targeting dysregulated cytokine secretion from neoplastic MKs as a novel therapeutic avenue. Unconventional secretion of TGFβ1 as well as interleukin 1β (IL1β) via secretory autophagy occurs in cells other than MKs, which prompted us to investigate whether similar mechanisms are utilized by MKs. Here, we identified that TGFβ1 strongly co-localized with the autophagy marker light chain 3B in native MKs. Disrupting secretory autophagy by inhibiting the small GTPase RhoA or its downstream effector Rho kinase (ROCK) markedly reduced TGFβ1 and IL1β secretion in vitro . In vivo , conditional deletion of the essential autophagy gene Atg5 from the hematopoietic system limited megakaryocytosis and aberrant cytokine secretion in an MPL W515L -driven transplant model. Similarly, mice with a selective deletion of Rhoa from the MK and platelet lineage were protected from progressive fibrosis. Finally, disease hallmarks in MPL W515L -transplanted mice were attenuated upon treatment with the autophagy inhibitor hydroxychloroquine or the ROCK inhibitor Y27632, either as monotherapy or in combination with the JAK2 inhibitor ruxolitinib. Overall, our data indicate that aberrant cytokine secretion is dependent on secretory autophagy downstream of RhoA, targeting of which represents a novel therapeutic avenue in the treatment of myelofibrosis. One Sentence Summary TGFβ1 is released from megakaryocytes via RhoA-mediated secretory autophagy, and targeting this process can alleviate fibrosis progression in a preclinical mouse model of myelofibrosis.
Collapse
|
4
|
Xiu Y, Xiong M, Yang H, Wang Q, Zhao X, Long J, Liang F, Liu N, Chen F, Gao M, Sun Y, Fan R, Zeng Y. Proteomic characterization of murine hematopoietic stem progenitor cells reveals dynamic fetal-to-adult changes in metabolic-related pathways. Biochem Biophys Res Commun 2024; 734:150661. [PMID: 39243675 DOI: 10.1016/j.bbrc.2024.150661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/24/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Hematopoietic stem progenitor cells (HSPCs) give rise to the hematopoietic system, maintain hematopoiesis throughout the lifespan, and undergo molecular and functional changes during their development and aging. The importance of hematopoietic stem cell (HSC) biology has led to their extensive characterization at genomic and transcriptomic levels. However, the proteomics of HSPCs throughout the murine lifetime still needs to be fully completed. Here, using mass spectrometry (MS)-based quantitative proteomics, we report on the dynamic changes in the proteome of HSPCs from four developmental stages in the fetal liver (FL) and the bone marrow (BM), including E14.5, young (2 months), middle-aged (8 months), and aging (18 months) stages. Proteomics unveils highly dynamic protein kinetics during the development and aging of HSPCs. Our data identify stage-specific developmental features of HSPCs, which can be linked to their functional maturation and senescence. Our proteomic data demonstrated that FL HSPCs depend on aerobic respiration to meet their proliferation and oxygen supply demand, while adult HSPCs prefer glycolysis to preserve the HSC pool. By functional assays, we validated the decreased mitochondrial metabolism, glucose uptake, reactive oxygen species (ROS) production, protein synthesis rate, and increased glutathione S-transferase (GST) activity during HSPC development from fetal to adult. Distinct metabolism pathways and immune-related pathways enriched in different HSPC developmental stages were revealed at the protein level. Our study will have broader implications for understanding the mechanism of stem cell maintenance and fate determination and reversing the HSC aging process.
Collapse
Affiliation(s)
- Yanyu Xiu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China; Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Mingfang Xiong
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China; Medical School of the Chinese PLA General Hospital, Beijing, 100039, China
| | - Haoyu Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China; Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Qianqian Wang
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China; School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Xiao Zhao
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Juan Long
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Fei Liang
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Nan Liu
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Fudong Chen
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China
| | - Meng Gao
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Yuying Sun
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Ruiwen Fan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China.
| | - Yang Zeng
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China; Medical School of the Chinese PLA General Hospital, Beijing, 100039, China; School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China.
| |
Collapse
|
5
|
Phillips E, van Enk S, Kildgaard S, Schlue S, Göttmann M, Jennings V, Bethke F, Müller G, Herold-Mende C, Pastor-Flores D, Schneider M, Helm D, Ostenfeld Larsen T, Goidts V. Malformin C preferentially kills glioblastoma stem-like cells via concerted induction of proteotoxic stress and autophagic flux blockade. Mol Oncol 2024. [PMID: 39462997 DOI: 10.1002/1878-0261.13756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/12/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024] Open
Abstract
Glioblastoma is a highly aggressive brain tumor for which there is no cure. The dire prognosis of this disease is largely attributable to a high level of heterogeneity, including the presence of a subpopulation of tumor-initiating glioblastoma stem-like cells (GSCs), which are refractory to chemo- and radiotherapy. Here, in an unbiased marine-derived fungal extract screen, together with bioguided dereplication based on high-resolution mass spectrometry, we identified malformin C to preferentially induce cell death in patient-derived GSCs and explore the potential of this cyclic peptide as a therapeutic agent for glioblastoma. Malformin C significantly reduced tumor growth in an in vivo xenograft model of glioblastoma. Using transcriptomics and chemoproteomics, we found that malformin C binds to many proteins, leading to their aggregation, and rapidly induces the unfolded protein response, including autophagy, in GSCs. Crucially, chemical inhibition of translation using cycloheximide rescued malformin C-induced cell death in GSCs, demonstrating that the proteotoxic effect of the compound is necessary for its cytotoxicity. At the same time, malformin C appears to accumulate in lysosomes, disrupting autophagic flux, and driving cells to death. Supporting this, malformin C synergizes with chloroquine, an inhibitor of autophagy. Strikingly, we observed that autophagic flux is differentially regulated in GSCs compared with normal astrocytes. The sensitivity of GSCs to malformin C highlights the relevance of proteostasis and autophagy as a therapeutic vulnerability in glioblastoma.
Collapse
Affiliation(s)
- Emma Phillips
- Junior Research Group "Brain Tumor Translational Targets", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sizèd van Enk
- Junior Research Group "Brain Tumor Translational Targets", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sara Kildgaard
- Department of Biotechnology and Biomedicine, Section for Microbial and Chemical Ecology, Natural Product Discovery, Technical University of Denmark, Copenhagen, Denmark
| | - Silja Schlue
- Junior Research Group "Brain Tumor Translational Targets", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mona Göttmann
- Junior Research Group "Brain Tumor Translational Targets", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Victoria Jennings
- Junior Research Group "Brain Tumor Translational Targets", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frederic Bethke
- Junior Research Group "Brain Tumor Translational Targets", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gabriele Müller
- Junior Research Group "Brain Tumor Translational Targets", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christel Herold-Mende
- Division of Neurosurgical Research, Department of Neurosurgery, University Hospital Heidelberg, Germany
| | - Daniel Pastor-Flores
- Division of Redox Regulation, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Schneider
- Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominic Helm
- Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Ostenfeld Larsen
- Department of Biotechnology and Biomedicine, Section for Microbial and Chemical Ecology, Natural Product Discovery, Technical University of Denmark, Copenhagen, Denmark
| | - Violaine Goidts
- Junior Research Group "Brain Tumor Translational Targets", German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
6
|
Man CH, Li C, Xu X, Zhao M. Metabolic regulation in normal and leukemic stem cells. Trends Pharmacol Sci 2024; 45:919-930. [PMID: 39306527 DOI: 10.1016/j.tips.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 10/06/2024]
Abstract
Hematopoietic stem cells (HSCs) and leukemic stem cells (LSCs) are crucial for ensuring hematopoietic homeostasis and driving leukemia progression, respectively. Recent research has revealed that metabolic adaptations significantly regulate the function and survival of these stem cells. In this review, we provide an overview of how metabolic pathways regulate oxidative and proteostatic stresses in HSCs during homeostasis and aging. Furthermore, we highlight targetable metabolic pathways and explore their interactions with epigenetics and the microenvironment in addressing the chemoresistance and immune evasion capacities of LSCs. The metabolic differences between HSCs and LSCs have profound implications for therapeutic strategies.
Collapse
Affiliation(s)
- Cheuk-Him Man
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Changzheng Li
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xi Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510030, China
| | - Meng Zhao
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
7
|
Lam K, Kim YJ, Ong CM, Liu AZ, Zhou FJ, Sunshine MJ, Chua BA, Vicenzi S, Ford PW, Zhou JH, Hong Y, Bennett EJ, Crews LA, Ball ED, Signer RAJ. The Proteostasis Network is a Therapeutic Target in Acute Myeloid Leukemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614781. [PMID: 39386464 PMCID: PMC11463481 DOI: 10.1101/2024.09.24.614781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Oncogenic growth places great strain and dependence on the proteostasis network. This has made proteostasis pathways attractive therapeutic targets in cancer, but efforts to drug these pathways have yielded disappointing clinical outcomes. One exception is proteasome inhibitors, which are approved for frontline treatment of multiple myeloma. However, proteasome inhibitors are largely ineffective for treatment of other cancers, including acute myeloid leukemia (AML), although reasons for these differences are unknown. Here, we determined that proteasome inhibitors are ineffective in AML due to inability to disrupt proteostasis. In response to proteasome inhibition, AML cells activated HSF1 and autophagy, two key stem cell proteostasis pathways, to prevent unfolded protein accumulation. Inactivation of HSF1 sensitized human AML cells to proteasome inhibition, marked by unfolded protein accumulation, activation of the PERK-mediated integrated stress response, severe reductions in protein synthesis, proliferation and cell survival, and significant slowing of disease progression and extension of survival in vivo . Similarly, combined autophagy and proteasome inhibition suppressed proliferation, synergistically killed AML cells, and significantly reduced AML burden and extended survival in vivo . Furthermore, autophagy and proteasome inhibition preferentially suppressed protein synthesis and induced apoptosis in primary patient AML cells, including AML stem/progenitor cells, without severely affecting normal hematopoietic stem/progenitor cells. Combined autophagy and proteasome inhibition also activated the integrated stress response, but surprisingly this occurred in a PKR-dependent manner. These studies unravel how proteostasis pathways are co-opted to promote AML growth, progression and drug resistance, and reveal that disabling the proteostasis network is a promising strategy to therapeutically target AML.
Collapse
|
8
|
Xiong M, Xiu Y, Long J, Zhao X, Wang Q, Yang H, Yu H, Bian L, Ju Y, Yin H, Hou Q, Liang F, Liu N, Chen F, Fan R, Sun Y, Zeng Y. Proteomics reveals dynamic metabolic changes in human hematopoietic stem progenitor cells from fetal to adulthood. Stem Cell Res Ther 2024; 15:303. [PMID: 39278906 PMCID: PMC11403967 DOI: 10.1186/s13287-024-03930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/06/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Hematopoietic stem progenitor cells (HSPCs) undergo phenotypical and functional changes during their emergence and development. Although the molecular programs governing the development of human hematopoietic stem cells (HSCs) have been investigated broadly, the relationships between dynamic metabolic alterations and their functions remain poorly characterized. METHODS In this study, we comprehensively described the proteomics of HSPCs in the human fetal liver (FL), umbilical cord blood (UCB), and adult bone marrow (aBM). The metabolic state of human HSPCs was assessed via a Seahorse assay, RT‒PCR, and flow cytometry-based metabolic-related analysis. To investigate whether perturbing glutathione metabolism affects reactive oxygen species (ROS) production, the metabolic state, and the expansion of human HSPCs, HSPCs were treated with buthionine sulfoximine (BSO), an inhibitor of glutathione synthetase, and N-acetyl-L-cysteine (NAC). RESULTS We investigated the metabolomic landscape of human HSPCs from the fetal, perinatal, and adult developmental stages by in-depth quantitative proteomics and predicted a metabolic switch from the oxidative state to the glycolytic state during human HSPC development. Seahorse assays, mitochondrial activity, ROS level, glucose uptake, and protein synthesis rate analysis supported our findings. In addition, immune-related pathways and antigen presentation were upregulated in UCB or aBM HSPCs, indicating their functional maturation upon development. Glutathione-related metabolic perturbations resulted in distinct responses in human HSPCs and progenitors. Furthermore, the molecular and immunophenotypic differences between human HSPCs at different developmental stages were revealed at the protein level for the first time. CONCLUSION The metabolic landscape of human HSPCs at three developmental stages (FL, UCB, and aBM), combined with proteomics and functional validations, substantially extends our understanding of HSC metabolic regulation. These findings provide valuable resources for understanding human HSC function and development during fetal and adult life.
Collapse
Affiliation(s)
- Mingfang Xiong
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China
| | - Yanyu Xiu
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Juan Long
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Xiao Zhao
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Qianqian Wang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Haoyu Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Hang Yu
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China
| | - Lihong Bian
- Department of Gynecology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Yan Ju
- Department of Gynecology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Hongyu Yin
- Department of Gynecology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Qingxiang Hou
- Department of Obstetrics and Gynecology, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Fei Liang
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Nan Liu
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Fudong Chen
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China
| | - Ruiwen Fan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Yuying Sun
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
| | - Yang Zeng
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China.
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China.
| |
Collapse
|
9
|
Su J, Wang Y, Yao J, Sun L, Zhao C, Liu L, Zhang L. Systemic knockout of Tmem175 results in aberrant differentiation but no effect on hematopoietic reconstitution. Stem Cell Res 2024; 79:103469. [PMID: 38878670 DOI: 10.1016/j.scr.2024.103469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/06/2024] [Accepted: 06/09/2024] [Indexed: 08/04/2024] Open
Abstract
Lysosomes play crucial roles in regulating cell metabolism, and K+ channels are critical for controlling various aspects of lysosomal function. Additionally, lysosomal activity is essential for maintaining the quiescence of hematopoietic stem cells (HSCs) under both steady-state and stress conditions. Tmem175 is a lysosomal potassium channel protein. To further investigate the role of K+ channels in HSCs, our study employed knockout mice to examine the function of Tmem175. Our research findings demonstrate that the deletion of Tmem175 does not disrupt the functionality of HSCs in both stable and stressed conditions, including irradiation and intraperitoneal 5-FU injections. However, we did observe that the absence of Tmem175 impairs the long-term differentiation capacity of HSCs into myeloid differentiated subpopulation cells(In this paper, it is referred to simply as M cells)in HSC transplantation test, while promoting their differentiation into T cells. This suggests that Tmem175 plays a role in the lineage differentiation of HSCs without being essential for their self-renewal or long-term regenerative capabilities.
Collapse
Affiliation(s)
- Jingjing Su
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Shandong Second Medical University, Weifang , 261053, China
| | - Yue Wang
- Center for Metabolic Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jiyuan Yao
- Center for Metabolic Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Leimin Sun
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Chunzhen Zhao
- Key Laboratory of Molecular Pharmacology and Translational Medicine and Department of Pharmacology, College of Pharmacy, Shandong Second Medical University, Weifang , 261053, China.
| | - Leiming Liu
- Key Laboratory of Artificial Organs and Computational Medicine, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, 310015, China.
| | - Lingling Zhang
- Center for Metabolic Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
10
|
Yang L, Shi J, Zhong M, Sun P, Zhang X, Lian Z, Yin H, Xu L, He G, Xu H, Wu H, Wang Z, Miao K, Huang J. NXPH4 mediated by m 5C contributes to the malignant characteristics of colorectal cancer via inhibiting HIF1A degradation. Cell Mol Biol Lett 2024; 29:111. [PMID: 39164641 PMCID: PMC11334498 DOI: 10.1186/s11658-024-00630-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 08/08/2024] [Indexed: 08/22/2024] Open
Abstract
OBJECTIVE Colorectal cancer (CRC) is a form of malignancy that exhibits a comparatively elevated occurrence and fatality rate. Given the relatively slower progress in diagnostic and therapeutic approaches for CRC, there is a need to investigate more accurate and efficient biomarkers. METHODS Core regulatory genes were screened using the TCGA database, and the expression of neurexophilin 4 (NXPH4) and its prognostic implications were validated using tissue microarray staining. The assessment of NXPH4 functions involved a range of experiments, including cellular, organoid, and murine models. Furthermore, a regulatory network between m5C, NXPH4, and HIF1A was established through several in vitro experiments. RESULTS The overexpression of NXPH4 is associated with unfavorable prognoses in patients with CRC and hepatocellular carcinoma. Additionally, it facilitates the progression of malignant tumors both in laboratory settings and in living organisms of colorectal carcinoma. Our research also reveals that NXPH4 mRNA can avoid degradation through RNautophagy, relying on an m5C-dependent mechanism. Moreover, NXPH4 amplifies the HIF signaling pathway and stabilizes HIF1A by competitively binding to PHD4. CONCLUSIONS NXPH4, regulated by m5C, promotes malignant tumor progression and regulates the HIF pathway. Consequently, targeting NXPH4 through molecular therapies could potentially serve as an efficacious therapeutic strategy for the management of CRC exhibiting elevated NXPH4 expression.
Collapse
Affiliation(s)
- Lei Yang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Department of Oncology, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
- Department of Clinical Biobank and Institute of Oncology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Jiawen Shi
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Department of Oncology, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Mingyang Zhong
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Department of Oncology, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Pingping Sun
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Department of Oncology, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
- Department of Clinical Biobank and Institute of Oncology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xiaojing Zhang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Department of Oncology, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
- Department of Clinical Biobank and Institute of Oncology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Zhengyi Lian
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Department of Oncology, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Hang Yin
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Department of Oncology, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Lijun Xu
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Department of Oncology, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Guyin He
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Department of Oncology, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Haiyan Xu
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Department of Oncology, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Han Wu
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Ziheng Wang
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, 999078, China
| | - Kai Miao
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, 999078, China.
| | - Jianfei Huang
- Clinical and Translational Research Center, Affiliated Hospital of Nantong University, Department of Oncology, Medical School of Nantong University, Nantong, 226001, Jiangsu, China.
- Department of Clinical Biobank and Institute of Oncology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
11
|
Pek JW. The idiosyncrasies of oocytes. Trends Cell Biol 2024:S0962-8924(24)00144-2. [PMID: 39142921 DOI: 10.1016/j.tcb.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024]
Abstract
Animal oocytes face extreme challenges. They remain dormant in the body for long periods of time. To support offspring development and health, they need to store genetic material and maternal factors stably and at the same time manage cellular damage in a reliable manner. Recent studies have provided new insights on how oocytes cope with such challenges. This review discusses the many unusual or idiosyncratic nature of oocytes and how understanding oocyte biology can help us address issues of reproduction and intergenerational inheritance.
Collapse
Affiliation(s)
- Jun Wei Pek
- Temasek Life Sciences Laboratory, 1 Research Link National University of Singapore, Singapore 117604, Singapore; Department of Biological Sciences, National University of Singapore, 14 Science Drive, 117543, Singapore.
| |
Collapse
|
12
|
Watanuki S, Kobayashi H, Sugiura Y, Yamamoto M, Karigane D, Shiroshita K, Sorimachi Y, Morikawa T, Fujita S, Shide K, Haraguchi M, Tamaki S, Mikawa T, Kondoh H, Nakano H, Sumiyama K, Nagamatsu G, Goda N, Okamoto S, Nakamura-Ishizu A, Shimoda K, Suematsu M, Suda T, Takubo K. SDHAF1 confers metabolic resilience to aging hematopoietic stem cells by promoting mitochondrial ATP production. Cell Stem Cell 2024; 31:1145-1161.e15. [PMID: 38772377 DOI: 10.1016/j.stem.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 02/20/2024] [Accepted: 04/30/2024] [Indexed: 05/23/2024]
Abstract
Aging generally predisposes stem cells to functional decline, impairing tissue homeostasis. Here, we report that hematopoietic stem cells (HSCs) acquire metabolic resilience that promotes cell survival. High-resolution real-time ATP analysis with glucose tracing and metabolic flux analysis revealed that old HSCs reprogram their metabolism to activate the pentose phosphate pathway (PPP), becoming more resistant to oxidative stress and less dependent on glycolytic ATP production at steady state. As a result, old HSCs can survive without glycolysis, adapting to the physiological cytokine environment in bone marrow. Mechanistically, old HSCs enhance mitochondrial complex II metabolism during stress to promote ATP production. Furthermore, increased succinate dehydrogenase assembly factor 1 (SDHAF1) in old HSCs, induced by physiological low-concentration thrombopoietin (TPO) exposure, enables rapid mitochondrial ATP production upon metabolic stress, thereby improving survival. This study provides insight into the acquisition of resilience through metabolic reprogramming in old HSCs and its molecular basis to ameliorate age-related hematopoietic abnormalities.
Collapse
Affiliation(s)
- Shintaro Watanuki
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Division of Hematology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hiroshi Kobayashi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Department of Cell Fate Biology and Stem Cell Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan.
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan; Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Masamichi Yamamoto
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center, Osaka 564-8565, Japan
| | - Daiki Karigane
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Division of Hematology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kohei Shiroshita
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Division of Hematology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yuriko Sorimachi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Department of Life Sciences and Medical BioScience, Waseda University School of Advanced Science and Engineering, Tokyo 162-8480, Japan
| | - Takayuki Morikawa
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Shinya Fujita
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Division of Hematology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kotaro Shide
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Miho Haraguchi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Shinpei Tamaki
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Takumi Mikawa
- Geriatric Unit, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Hiroshi Kondoh
- Geriatric Unit, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Hiroyasu Nakano
- Department of Biochemistry, Toho University School of Medicine, Tokyo 143-8540, Japan
| | - Kenta Sumiyama
- Laboratory of Animal Genetics and Breeding, Department of Animal Sciences, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan; RIKEN Center for Biosystems Dynamics Research, Laboratory for Mouse Genetic Engineering, Osaka 565-0871, Japan
| | - Go Nagamatsu
- Center for Advanced Assisted Reproductive Technologies, University of Yamanashi, Kofu 400-8501, Japan
| | - Nobuhito Goda
- Department of Life Sciences and Medical BioScience, Waseda University School of Advanced Science and Engineering, Tokyo 162-8480, Japan
| | - Shinichiro Okamoto
- Division of Hematology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Ayako Nakamura-Ishizu
- Department of Microscopic and Developmental Anatomy, Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | - Kazuya Shimoda
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan; Live Imaging Center, Central Institute for Experimental Medicine and Life Science, Kawasaki 210-0821, Japan
| | - Toshio Suda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Keiyo Takubo
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Department of Cell Fate Biology and Stem Cell Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan.
| |
Collapse
|
13
|
Larrue C, Mouche S, Angelino P, Sajot M, Birsen R, Kosmider O, Mckee T, Vergez F, Recher C, Mas VMD, Gu Q, Xu J, Tsantoulis P, Sarry JE, Tamburini J. Targeting ferritinophagy impairs quiescent cancer stem cells in acute myeloid leukemia in vitro and in vivo models. Sci Transl Med 2024; 16:eadk1731. [PMID: 39047119 DOI: 10.1126/scitranslmed.adk1731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 03/18/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024]
Abstract
Acute myeloid leukemia (AML) remains a challenging hematological malignancy with poor prognosis and limited treatment options. Leukemic stem cells (LSCs) contribute to therapeutic failure, relapse, and adverse outcome. This study investigates the role of quiescence and related molecular mechanisms in AML pathogenesis and LSC functions to identify potential therapeutic targets. Transcriptomic analysis revealed that the LSC-enriched quiescent cell population has a distinct gene signature with prognostic relevance in patients with AML. Mechanistically, quiescent blasts exhibit increased autophagic activity, which contributes to their sustained viability. Proteomic profiling uncovered differential requirements for iron metabolism between quiescent and cycling cells, revealing a unique dependence of quiescent cells on ferritinophagy, a selective form of autophagy mediated by nuclear receptor coactivator 4 (NCOA4), which regulates iron bioavailability. We evaluated the therapeutic potential of inhibiting NCOA4-mediated ferritinophagy using genetic knockdown and chemical inhibition approaches. In vitro assays showed that suppression of NCOA4 was toxic to leukemic blasts, particularly the CD34+CD38- LSC-enriched population, without affecting normal CD34+ hematopoietic progenitors. In vivo studies using murine patient-derived xenograft (PDX) models of AML confirmed that NCOA4 inhibition reduced tumor burden and impaired LSC viability and self-renewal, indicating a specific vulnerability of these cells to ferritinophagy disruption. Our findings underscore the role of NCOA4-mediated ferritinophagy in maintaining LSC quiescence and function and suggest that targeting this pathway may be an effective therapeutic strategy for AML. This study highlights the potential of NCOA4 inhibition to improve AML outcomes and paves the way for future research and clinical development.
Collapse
Affiliation(s)
- Clement Larrue
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, 31100 Toulouse, France
- LabEx Toucan, 31100 Toulouse, France
- Équipe labellisée Ligue Nationale Contre le Cancer 2023, 31100 Toulouse, France
| | - Sarah Mouche
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
| | - Paolo Angelino
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
- Translational Data Science, SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Maxime Sajot
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, 31100 Toulouse, France
- LabEx Toucan, 31100 Toulouse, France
- Équipe labellisée Ligue Nationale Contre le Cancer 2023, 31100 Toulouse, France
| | - Rudy Birsen
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, 75014 Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, 75014 Paris, France
| | - Olivier Kosmider
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, 75014 Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, 75014 Paris, France
| | - Thomas Mckee
- Division of Clinical Pathology, Diagnostic Department, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - François Vergez
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, 31100 Toulouse, France
| | - Christian Recher
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, 31100 Toulouse, France
| | - Véronique Mansat-De Mas
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Service d'Hématologie, 31100 Toulouse, France
| | - Qiong Gu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jun Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Petros Tsantoulis
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm U1037, CNRS U5077, 31100 Toulouse, France
- LabEx Toucan, 31100 Toulouse, France
- Équipe labellisée Ligue Nationale Contre le Cancer 2023, 31100 Toulouse, France
| | - Jerome Tamburini
- Centre for Translational Research in Onco-Hematology, Faculty of Medicine, University of Geneva, and Swiss Cancer Centre Leman, 1206 Geneva, Switzerland
- Université de Paris, Institut Cochin, CNRS U8104, Inserm U1016, 75014 Paris, France
- Oncology Department, Geneva University Hospitals, 1205 Geneva, Switzerland
| |
Collapse
|
14
|
Zuo H, Wu A, Wang M, Hong L, Wang H. tRNA m 1A modification regulate HSC maintenance and self-renewal via mTORC1 signaling. Nat Commun 2024; 15:5706. [PMID: 38977676 PMCID: PMC11231335 DOI: 10.1038/s41467-024-50110-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/28/2024] [Indexed: 07/10/2024] Open
Abstract
Haematopoietic stem cells (HSCs) possess unique physiological adaptations to sustain blood cell production and cope with stress responses throughout life. To maintain these adaptations, HSCs rely on maintaining a tightly controlled protein translation rate. However, the mechanism of how HSCs regulate protein translation remains to be fully elucidated. In this study, we investigate the role of transfer RNA (tRNA) m1A58 'writer' proteins TRMT6 and TRMT61A in regulating HSCs function. Trmt6 deletion promoted HSC proliferation through aberrant activation of mTORC1 signaling. TRMT6-deficient HSCs exhibited an impaired self-renewal ability in competitive transplantation assay. Mechanistically, single cell RNA-seq analysis reveals that the mTORC1 signaling pathway is highly upregulated in HSC-enriched cell populations after Trmt6 deletion. m1A-tRNA-seq and Western blot analysis suggest that TRMT6 promotes methylation modification of specific tRNA and expression of TSC1, fine-tuning mTORC1 signaling levels. Furthermore, Pharmacological inhibition of the mTORC1 pathway rescued functional defect in TRMT6-deficient HSCs. To our knowledge, this study is the first to elucidate a mechanism by which TRMT6-TRMT61A complex-mediated tRNA-m1A58 modification regulates HSC homeostasis.
Collapse
Affiliation(s)
- Hongna Zuo
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, China
| | - Aiwei Wu
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, China
| | - Mingwei Wang
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, China
| | - Liquan Hong
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, China
| | - Hu Wang
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People's Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, China.
| |
Collapse
|
15
|
Yokomizo T, Oshima M, Iwama A. Epigenetics of hematopoietic stem cell aging. Curr Opin Hematol 2024; 31:207-216. [PMID: 38640057 DOI: 10.1097/moh.0000000000000818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
PURPOSE OF REVIEW The development of new antiaging medicines is of great interest to the current elderly and aging population. Aging of the hematopoietic system is attributed to the aging of hematopoietic stem cells (HSCs), and epigenetic alterations are the key effectors driving HSC aging. Understanding the epigenetics of HSC aging holds promise of providing new insights for combating HSC aging and age-related hematological malignancies. RECENT FINDINGS Aging is characterized by the progressive loss of physiological integrity, leading to impaired function and increased vulnerability to death. During aging, the HSCs undergo both quantitative and qualitative changes. These functional changes in HSCs cause dysregulated hematopoiesis, resulting in anemia, immune dysfunction, and an increased risk of hematological malignancies. Various cell-intrinsic and cell-extrinsic effectors influencing HSC aging have also been identified. Epigenetic alterations are one such mechanism. SUMMARY Cumulative epigenetic alterations in aged HSCs affect their fate, leading to aberrant self-renewal, differentiation, and function of aged HSCs. In turn, these factors provide an opportunity for aged HSCs to expand by modulating their self-renewal and differentiation balance, thereby contributing to the development of hematological malignancies.
Collapse
Affiliation(s)
- Takako Yokomizo
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|
16
|
Aguadé-Gorgorió J, Jami-Alahmadi Y, Calvanese V, Kardouh M, Fares I, Johnson H, Rezek V, Ma F, Magnusson M, Wang Y, Shin JE, Nance KJ, Goodridge HS, Liebscher S, Schenke-Layland K, Crooks GM, Wohlschlegel JA, Mikkola HKA. MYCT1 controls environmental sensing in human haematopoietic stem cells. Nature 2024; 630:412-420. [PMID: 38839950 PMCID: PMC11168926 DOI: 10.1038/s41586-024-07478-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 04/26/2024] [Indexed: 06/07/2024]
Abstract
The processes that govern human haematopoietic stem cell (HSC) self-renewal and engraftment are poorly understood and challenging to recapitulate in culture to reliably expand functional HSCs1-3. Here we identify MYC target 1 (MYCT1; also known as MTLC) as a crucial human HSC regulator that moderates endocytosis and environmental sensing in HSCs. MYCT1 is selectively expressed in undifferentiated human haematopoietic stem and progenitor cells (HSPCs) and endothelial cells but becomes markedly downregulated during HSC culture. Lentivirus-mediated knockdown of MYCT1 prevented human fetal liver and cord blood (CB) HSPC expansion and engraftment. By contrast, restoring MYCT1 expression improved the expansion and engraftment of cultured CB HSPCs. Single-cell RNA sequencing of human CB HSPCs in which MYCT1 was knocked down or overexpressed revealed that MYCT1 governs important regulatory programmes and cellular properties essential for HSC stemness, such as ETS factor expression and low mitochondrial activity. MYCT1 is localized in the endosomal membrane in HSPCs and interacts with vesicle trafficking regulators and signalling machinery. MYCT1 loss in HSPCs led to excessive endocytosis and hyperactive signalling responses, whereas restoring MYCT1 expression balanced culture-induced endocytosis and dysregulated signalling. Moreover, sorting cultured CB HSPCs on the basis of lowest endocytosis rate identified HSPCs with preserved MYCT1 expression and MYCT1-regulated HSC stemness programmes. Our work identifies MYCT1-moderated endocytosis and environmental sensing as essential regulatory mechanisms required to preserve human HSC stemness. Our data also pinpoint silencing of MYCT1 as a cell-culture-induced vulnerability that compromises human HSC expansion.
Collapse
Affiliation(s)
- Júlia Aguadé-Gorgorió
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA.
| | - Yasaman Jami-Alahmadi
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA, USA
- Pfizer, Cambridge, MA, USA
| | - Vincenzo Calvanese
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA
- Laboratory for Molecular Cell Biology, University College London, London, UK
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Maya Kardouh
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
- Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | - Iman Fares
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
- Kite Pharma, Santa Monica, CA, USA
| | - Haley Johnson
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Valerie Rezek
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- UCLA AIDS Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Feiyang Ma
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
- Institute for Genomics and Proteomics, University of California Los Angeles, Los Angeles, CA, USA
- Amgen, Thousand Oaks, CA, USA
| | - Mattias Magnusson
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Yanling Wang
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Juliana E Shin
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Karina J Nance
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Helen S Goodridge
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Simone Liebscher
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University, Tübingen, Germany
| | - Katja Schenke-Layland
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University Tübingen, Reutlingen, Germany
| | - Gay M Crooks
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - James A Wohlschlegel
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Hanna K A Mikkola
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Fleetwood AJ, Noonan J, La Gruta N, Kallies A, Murphy AJ. Immunometabolism in atherosclerotic disorders. NATURE CARDIOVASCULAR RESEARCH 2024; 3:637-650. [PMID: 39196223 DOI: 10.1038/s44161-024-00473-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 04/11/2024] [Indexed: 08/29/2024]
Abstract
Cardiovascular diseases (CVDs), including atherosclerosis, myocardial infarction and heart failure, are the leading causes of morbidity and mortality worldwide. Emerging evidence suggests a crucial role for immune cell dysfunction and inflammation in the progression of this complex set of diseases. Recent advances demonstrate that immune cells, tightly linked to CVD pathogenesis, are sensitive to environmental signals and respond by engaging immunometabolic networks that shape their behavior. Inflammatory cues and altered nutrient availability within atherosclerotic plaques or following ischemia synergize to elicit metabolic shifts in immune cells that influence the course of disease pathology. Understanding these metabolic adaptations and how they contribute to cellular dysfunction may reveal novel therapeutic approaches for the treatment of CVD. Here we provide a comprehensive summary of the metabolic reprogramming that occurs in immune cells and their progenitors during CVD, offering insights into the potential therapeutic interventions to mitigate disease progression.
Collapse
Affiliation(s)
- Andrew J Fleetwood
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
| | - Jonathan Noonan
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Nicole La Gruta
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Axel Kallies
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
| |
Collapse
|
18
|
Maneix L, Iakova P, Lee CG, Moree SE, Lu X, Datar GK, Hill CT, Spooner E, King JCK, Sykes DB, Saez B, Di Stefano B, Chen X, Krause DS, Sahin E, Tsai FTF, Goodell MA, Berk BC, Scadden DT, Catic A. Cyclophilin A supports translation of intrinsically disordered proteins and affects haematopoietic stem cell ageing. Nat Cell Biol 2024; 26:593-603. [PMID: 38553595 PMCID: PMC11021199 DOI: 10.1038/s41556-024-01387-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 02/23/2024] [Indexed: 04/11/2024]
Abstract
Loss of protein function is a driving force of ageing. We have identified peptidyl-prolyl isomerase A (PPIA or cyclophilin A) as a dominant chaperone in haematopoietic stem and progenitor cells. Depletion of PPIA accelerates stem cell ageing. We found that proteins with intrinsically disordered regions (IDRs) are frequent PPIA substrates. IDRs facilitate interactions with other proteins or nucleic acids and can trigger liquid-liquid phase separation. Over 20% of PPIA substrates are involved in the formation of supramolecular membrane-less organelles. PPIA affects regulators of stress granules (PABPC1), P-bodies (DDX6) and nucleoli (NPM1) to promote phase separation and increase cellular stress resistance. Haematopoietic stem cell ageing is associated with a post-transcriptional decrease in PPIA expression and reduced translation of IDR-rich proteins. Here we link the chaperone PPIA to the synthesis of intrinsically disordered proteins, which indicates that impaired protein interaction networks and macromolecular condensation may be potential determinants of haematopoietic stem cell ageing.
Collapse
Affiliation(s)
- Laure Maneix
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Polina Iakova
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Charles G Lee
- Department of BioSciences, Rice University, Houston, TX, USA
| | - Shannon E Moree
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Xuan Lu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Gandhar K Datar
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Cedric T Hill
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eric Spooner
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Jordon C K King
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - David B Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Borja Saez
- Center for Applied Medical Research, Hematology-Oncology Unit, Pamplona, Navarra, Spain
| | - Bruno Di Stefano
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Xi Chen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Daniela S Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Ergun Sahin
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Francis T F Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Margaret A Goodell
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Bradford C Berk
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - David T Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - André Catic
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA.
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
| |
Collapse
|
19
|
Li L, Huang J, Zhang S, Yao C, Chen Y, Wang H, Guo B. The Chaperone Protein Cct5 is Essential for Hematopoietic Stem Cell Maintenance. Stem Cell Rev Rep 2024; 20:769-778. [PMID: 38153635 DOI: 10.1007/s12015-023-10670-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 12/29/2023]
Abstract
Proper proteostasis is indispensable for the long-term maintenance of hematopoietic stem and progenitor cells (HSPCs). The TRiC/CCT (chaperonin-containing TCP-1) complex, a key constituent of cellular machinery facilitating accurate protein folding, has remained enigmatic in its specific function within HSPCs. Here we show that conditional knockout (KO) of Cct5 significantly impairs the maintenance of murine HSPCs. Primary and secondary transplantation experiments unequivocally demonstrate the incapacity of Cct5 KO HSPCs to reconstitute both myeloid and lymphoid lineage cells in recipient mice, highlighting the pivotal role of the TRiC/CCT complex in governing these cellular lineages. Furthermore, leveraging an integrated approach that merges a Protein-Protein Interaction (PPI) database with RNA sequencing (RNA-seq) data of HSPCs, our analysis reveals intricate interactions between Cct5 and vital transcription factors crucial for HSC homeostasis. Notably, Cct5 engages with MYC, PIAS1, TP53, ESR1, HOXA1, and JUN, intricately regulating the transcriptional landscape governing autophagy, myeloid differentiation, and cytoskeleton organization within HSPCs. Our study unveils the profound significance of TRiC/CCT complex-mediated proteostasis in orchestrating the maintenance and functionality of HSPCs.
Collapse
Affiliation(s)
- Linxi Li
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280, Chong-Qing South Road, West Bldg #2, Rm 804, Shanghai, 200025, China
| | - Jie Huang
- Children's Hospital, Fudan University, Minhang, Shanghai, China
| | - Suping Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280, Chong-Qing South Road, West Bldg #2, Rm 804, Shanghai, 200025, China
| | - Chunxu Yao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280, Chong-Qing South Road, West Bldg #2, Rm 804, Shanghai, 200025, China
| | - Yandan Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280, Chong-Qing South Road, West Bldg #2, Rm 804, Shanghai, 200025, China.
- Department of Hematology, Xin Hua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200090, China.
| | - Haitao Wang
- Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Bin Guo
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280, Chong-Qing South Road, West Bldg #2, Rm 804, Shanghai, 200025, China.
- Department of Hematology, Xin Hua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200090, China.
| |
Collapse
|
20
|
Jiang Y, Ning Y, Cheng S, Huang Y, Deng M, Chen C. Single-cell aggrephagy-related patterns facilitate tumor microenvironment intercellular communication, influencing osteosarcoma progression and prognosis. Apoptosis 2024; 29:521-535. [PMID: 38066392 DOI: 10.1007/s10495-023-01922-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 02/18/2024]
Abstract
Osteosarcoma, a common malignant tumor in children, has emerged as a major threat to the life and health of pediatric patients. Presently, there are certain limitations in the diagnosis and treatment methods for this disease, resulting in inferior therapeutic outcomes. Therefore, it is of great importance to study its pathogenesis and explore innovative approaches to diagnosis and treatment. In this study, a non-negative matrix decomposition method was employed to conduct a comprehensive investigation and analysis of aggregated autophagy-related genes within 331,394 single-cell samples of osteosarcoma. Through this study, we have elucidated the intricate communication patterns among various cells within the tumor microenvironment. Based on the classification of aggregated autophagy-related genes, we are not only able to more accurately predict patients' prognosis but also offer robust guidance for treatment strategies. The findings of this study hold promise for breakthroughs in the diagnosis and treatment of osteosarcoma, intervention of aggrephagy is expected to improve the survival rate and quality of life of osteosarcoma patients.
Collapse
Affiliation(s)
- Yunsheng Jiang
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| | - Yun Ning
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
| | - Shidi Cheng
- Department of Hematology, Army Medical Center of PLA, Chongqing, 400012, China
| | - Yinde Huang
- Department of Breast and Thyroid Surgery, Chongqing General Hospital, Chongqing, 401147, China
| | - Muhai Deng
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| | - Cheng Chen
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
21
|
Liu Y, Shi L, Chen Y, Luo S, Chen Y, Chen H, Lan W, Lu X, Cao Z, Ye Z, Li J, Yu B, Dzierzak E, Li Z. Autophagy regulates the maturation of hematopoietic precursors in the embryo. Nat Commun 2024; 15:2255. [PMID: 38490977 PMCID: PMC10943005 DOI: 10.1038/s41467-024-46453-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 02/28/2024] [Indexed: 03/18/2024] Open
Abstract
An understanding of the mechanisms regulating embryonic hematopoietic stem cell (HSC) development would facilitate their regeneration. The aorta-gonad-mesonephros region is the site for HSC production from hemogenic endothelial cells (HEC). While several distinct regulators are involved in this process, it is not yet known whether macroautophagy (autophagy) plays a role in hematopoiesis in the pre-liver stage. Here, we show that different states of autophagy exist in hematopoietic precursors and correlate with hematopoietic potential based on the LC3-RFP-EGFP mouse model. Deficiency of autophagy-related gene 5 (Atg5) specifically in endothelial cells disrupts endothelial to hematopoietic transition (EHT), by blocking the autophagic process. Using combined approaches, including single-cell RNA-sequencing (scRNA-seq), we have confirmed that Atg5 deletion interrupts developmental temporal order of EHT to further affect the pre-HSC I maturation, and that autophagy influences hemogenic potential of HEC and the formation of pre-HSC I likely via the nucleolin pathway. These findings demonstrate a role for autophagy in the formation/maturation of hematopoietic precursors.
Collapse
Affiliation(s)
- Yumin Liu
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Linjuan Shi
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yifan Chen
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Sifan Luo
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuehang Chen
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hongtian Chen
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wenlang Lan
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xun Lu
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhan Cao
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zehua Ye
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jinping Li
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bo Yu
- Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Elaine Dzierzak
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | - Zhuan Li
- Key Laboratory of Functional Proteomics of Guangdong Province, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
22
|
Chagraoui J, Girard S, Mallinger L, Mayotte N, Tellechea MF, Sauvageau G. KBTBD4-mediated reduction of MYC is critical for hematopoietic stem cell expansion upon UM171 treatment. Blood 2024; 143:882-894. [PMID: 38207291 DOI: 10.1182/blood.2023021342] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/13/2024] Open
Abstract
ABSTRACT Ex vivo expansion of hematopoietic stem cells (HSCs) is gaining importance for cell and gene therapy, and requires a shift from dormancy state to activation and cycling. However, abnormal or excessive HSC activation results in reduced self-renewal ability and increased propensity for myeloid-biased differentiation. We now report that activation of the E3 ligase complex CRL3KBTBD4 by UM171 not only induces epigenetic changes through CoREST1 degradation but also controls chromatin-bound master regulator of cell cycle entry and proliferative metabolism (MYC) levels to prevent excessive activation and maintain lympho-myeloid potential of expanded populations. Furthermore, reconstitution activity and multipotency of UM171-treated HSCs are specifically compromised when MYC levels are experimentally increased despite degradation of CoREST1.
Collapse
Affiliation(s)
- Jalila Chagraoui
- Molecular Genetics of Stem Cells Laboratory, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
| | - Simon Girard
- Molecular Genetics of Stem Cells Laboratory, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
| | - Laure Mallinger
- Molecular Genetics of Stem Cells Laboratory, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
| | - Nadine Mayotte
- Molecular Genetics of Stem Cells Laboratory, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
| | - Maria Florencia Tellechea
- Molecular Genetics of Stem Cells Laboratory, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
| | - Guy Sauvageau
- Molecular Genetics of Stem Cells Laboratory, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada
- Division of Hematology, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
- Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
23
|
Zaffagnini G, Cheng S, Salzer MC, Pernaute B, Duran JM, Irimia M, Schuh M, Böke E. Mouse oocytes sequester aggregated proteins in degradative super-organelles. Cell 2024; 187:1109-1126.e21. [PMID: 38382525 DOI: 10.1016/j.cell.2024.01.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 12/04/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024]
Abstract
Oocytes are among the longest-lived cells in the body and need to preserve their cytoplasm to support proper embryonic development. Protein aggregation is a major threat for intracellular homeostasis in long-lived cells. How oocytes cope with protein aggregation during their extended life is unknown. Here, we find that mouse oocytes accumulate protein aggregates in specialized compartments that we named endolysosomal vesicular assemblies (ELVAs). Combining live-cell imaging, electron microscopy, and proteomics, we found that ELVAs are non-membrane-bound compartments composed of endolysosomes, autophagosomes, and proteasomes held together by a protein matrix formed by RUFY1. Functional assays revealed that in immature oocytes, ELVAs sequester aggregated proteins, including TDP-43, and degrade them upon oocyte maturation. Inhibiting degradative activity in ELVAs leads to the accumulation of protein aggregates in the embryo and is detrimental for embryo survival. Thus, ELVAs represent a strategy to safeguard protein homeostasis in long-lived cells.
Collapse
Affiliation(s)
- Gabriele Zaffagnini
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Shiya Cheng
- Department of Meiosis, Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Marion C Salzer
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Barbara Pernaute
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Juan Manuel Duran
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Manuel Irimia
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Melina Schuh
- Department of Meiosis, Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37077 Göttingen, Germany
| | - Elvan Böke
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
24
|
Petersen M, Ebstrup E, Rodriguez E. Going through changes - the role of autophagy during reprogramming and differentiation. J Cell Sci 2024; 137:jcs261655. [PMID: 38393817 DOI: 10.1242/jcs.261655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024] Open
Abstract
Somatic cell reprogramming is a complex feature that allows differentiated cells to undergo fate changes into different cell types. This process, which is conserved between plants and animals, is often achieved via dedifferentiation into pluripotent stem cells, which have the ability to generate all other types of cells and tissues of a given organism. Cellular reprogramming is thus a complex process that requires extensive modification at the epigenetic and transcriptional level, unlocking cellular programs that allow cells to acquire pluripotency. In addition to alterations in the gene expression profile, cellular reprogramming requires rearrangement of the proteome, organelles and metabolism, but these changes are comparatively less studied. In this context, autophagy, a cellular catabolic process that participates in the recycling of intracellular constituents, has the capacity to affect different aspects of cellular reprogramming, including the removal of protein signatures that might hamper reprogramming, mitophagy associated with metabolic reprogramming, and the supply of energy and metabolic building blocks to cells that undergo fate changes. In this Review, we discuss advances in our understanding of the role of autophagy during cellular reprogramming by drawing comparisons between plant and animal studies, as well as highlighting aspects of the topic that warrant further research.
Collapse
Affiliation(s)
- Morten Petersen
- Department of Biology, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Elise Ebstrup
- Department of Biology, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Eleazar Rodriguez
- Department of Biology, University of Copenhagen, 2200 Copenhagen N, Denmark
| |
Collapse
|
25
|
Karimnia N, Harris J, Heazlewood SY, Cao B, Nilsson SK. Metabolic regulation of aged hematopoietic stem cells: key players and mechanisms. Exp Hematol 2023; 128:2-9. [PMID: 37778498 DOI: 10.1016/j.exphem.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Affiliation(s)
- Nazanin Karimnia
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - James Harris
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, Australia; School of Clinical Sciences, Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Australia
| | - Shen Y Heazlewood
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - Benjamin Cao
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, Australia.
| | - Susan K Nilsson
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, Australia.
| |
Collapse
|
26
|
Kasbekar M, Mitchell CA, Proven MA, Passegué E. Hematopoietic stem cells through the ages: A lifetime of adaptation to organismal demands. Cell Stem Cell 2023; 30:1403-1420. [PMID: 37865087 PMCID: PMC10842631 DOI: 10.1016/j.stem.2023.09.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/20/2023] [Accepted: 09/28/2023] [Indexed: 10/23/2023]
Abstract
Hematopoietic stem cells (HSCs), which govern the production of all blood lineages, transition through a series of functional states characterized by expansion during fetal development, functional quiescence in adulthood, and decline upon aging. We describe central features of HSC regulation during ontogeny to contextualize how adaptive responses over the life of the organism ultimately form the basis for HSC functional degradation with age. We particularly focus on the role of cell cycle regulation, inflammatory response pathways, epigenetic changes, and metabolic regulation. We then explore how the knowledge of age-related changes in HSC regulation can inform strategies for the rejuvenation of old HSCs.
Collapse
Affiliation(s)
- Monica Kasbekar
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY 10032, USA; Division of Hematology and Medical Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Carl A Mitchell
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - Melissa A Proven
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY 10032, USA.
| |
Collapse
|