1
|
Chiang YH, Emmrich S, Vannini N. Metabolic Alterations in HSCs during Aging and Leukemogenesis. Physiology (Bethesda) 2025; 40:0. [PMID: 40019828 DOI: 10.1152/physiol.00054.2024] [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: 11/20/2024] [Revised: 12/15/2024] [Accepted: 02/23/2025] [Indexed: 04/26/2025] Open
Abstract
Aging is a multifaceted process associated with a functional decline in cellular function over time, affecting all lifeforms. During the aging process, metabolism, a fundamental hallmark of life (1), is profoundly altered. In the context of hematopoiesis, the proper function of hematopoietic stem cells, at the apex of the blood system, is tightly linked to their energy metabolism, which in turn shapes hematopoietic output. Here, we review the latest developments in our understanding of the metabolic states and changes in aged hematopoietic stem cells, molecular players and pathways involved in aged hematopoietic stem cell metabolism, the consequences of perturbed metabolism on clonal hematopoiesis and leukemogenesis, and pharmacologic/genetic strategies to reverse or rejuvenate altered metabolic phenotypes.
Collapse
Affiliation(s)
- Yi-Hsuan Chiang
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Stephan Emmrich
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Nicola Vannini
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
2
|
Yvan-Charvet L, Barouillet T, Borowczyk C. Haematometabolism rewiring in atherosclerotic cardiovascular disease. Nat Rev Cardiol 2025; 22:414-430. [PMID: 39743562 DOI: 10.1038/s41569-024-01108-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/14/2024] [Indexed: 01/04/2025]
Abstract
Atherosclerotic cardiovascular diseases are the most frequent cause of death worldwide. The clinical complications of atherosclerosis are closely linked to the haematopoietic and immune systems, which maintain homeostatic functions and vital processes in the body. The nodes linking metabolism and inflammation are receiving increasing attention because they are inextricably linked to inflammatory manifestations of non-communicable diseases, including atherosclerosis. Although metabolism and inflammation are essential to survival and involve all tissues, we still know little about how these processes influence each other. In an effort to understand these mechanisms, in this Review we explore whether and how potent cardiovascular risk factors and metabolic modifiers of atherosclerosis influence the molecular and cellular machinery of 'haematometabolism' (metabolic-dependent haematopoietic stem cell skewing) and 'efferotabolism' (metabolic-dependent efferocyte reprogramming). These changes might ultimately propagate a quantitative and qualitative drift of the macrophage supply chain and affect the clinical manifestations of atherosclerosis. Refining our understanding of the different metabolic requirements of these processes could open the possibility of developing therapeutics targeting haematometabolism that, in conjunction with improved dietary habits, help rebalance and promote efficient haematopoiesis and efferocytosis and decrease the risk of atherosclerosis complications.
Collapse
Affiliation(s)
- Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France.
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France.
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| | - Thibault Barouillet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | - Coraline Borowczyk
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France.
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France.
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| |
Collapse
|
3
|
Chaudhary JK, Danga AK, Kumari A, Bhardwaj A, Rath PC. Role of stem cells in ageing and age-related diseases. Mech Ageing Dev 2025; 225:112069. [PMID: 40324541 DOI: 10.1016/j.mad.2025.112069] [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: 01/17/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Stem cell functions and ageing are deeply interconnected, continually influencing each other in multiple ways. Stem cells play a vital role in organ maintenance, regeneration, and homeostasis, all of which decline over time due to gradual reduction in their self-renewal, differentiation, and growth factor secretion potential. The functional decline is attributed to damaging extrinsic environmental factors and progressively worsening intrinsic genetic and biochemical processes. These ageing-associated deteriorative changes have been extensively documented, paving the way for the discovery of novel biomarkers of ageing for detection, diagnosis, and treatment of age-related diseases. Age-dependent changes in adult stem cells include numerical decline, loss of heterogeneity, and reduced self-renewal and differentiation, leading to a drastic reduction in regenerative potential and thereby driving the ageing process. Conversely, ageing also adversely alters the stem cell niche, disrupting the molecular pathways underlying stem cell homing, self-renewal, differentiation, and growth factor secretion, all of which are critical for tissue repair and regeneration. A holistic understanding of these molecular mechanisms, through empirical research and clinical trials, is essential for designing targeted therapies to modulate ageing and improve health parameters in older individuals.
Collapse
Affiliation(s)
- Jitendra Kumar Chaudhary
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India; Department of Zoology, Shivaji College, University of Delhi, New Delhi 110027, India.
| | - Ajay Kumar Danga
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India; National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Anita Kumari
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Akshay Bhardwaj
- Global Research Alliances, Ashoka University, Rajiv Gandhi Education City, Sonepat, Haryana 131029, India.
| | - Pramod C Rath
- Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
4
|
Marchand T, Akinnola KE, Takeishi S, Maryanovich M, Pinho S, Saint-Vanne J, Birbrair A, Lamy T, Tarte K, Frenette P, Gritsman K. Periosteal skeletal stem cells can migrate into the bone marrow and support hematopoiesis after injury. eLife 2025; 13:RP101714. [PMID: 40401637 PMCID: PMC12097789 DOI: 10.7554/elife.101714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Abstract
Skeletal stem cells (SSCs) have been isolated from various tissues, including periosteum and bone marrow, where they exhibit key functions in bone biology and hematopoiesis, respectively. The role of periosteal SSCs (P-SSCs) in bone regeneration and healing has been extensively studied, but their ability to contribute to the bone marrow stroma is still under debate. In the present study, we characterized a mouse whole bone transplantation model that mimics the initial bone marrow necrosis and fatty infiltration seen after injury. Using this model and a lineage tracing approach, we observed the migration of P-SSCs into the bone marrow after transplantation. Once in the bone marrow, P-SSCs are phenotypically and functionally reprogrammed into bone marrow mesenchymal stem cells (BM-MSCs) that express high levels of hematopoietic stem cell niche factors such as Cxcl12 and Kitl. In addition, using ex vivo and in vivo approaches, we found that P-SSCs are more resistant to acute stress than BM-MSCs. These results highlight the plasticity of P-SSCs and their potential role in bone marrow regeneration after bone marrow injury.
Collapse
Affiliation(s)
- Tony Marchand
- Service d’hématologie Clinique, Centre Hospitalier Universitaire de RennesRennesFrance
- UMR U1236, INSERM, Université Rennes, EFS Bretagne, Equipe Labellisée Ligue Contre le CancerRennesFrance
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
| | - Kemi E Akinnola
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
| | - Shoichiro Takeishi
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
| | - Maria Maryanovich
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
| | - Sandra Pinho
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Medical Oncology, Albert Einstein College of MedicineBronxUnited States
- Department of Pharmacology & Regenerative Medicine, University of Illinois at ChicagoChicagoUnited States
| | - Julien Saint-Vanne
- UMR U1236, INSERM, Université Rennes, EFS Bretagne, Equipe Labellisée Ligue Contre le CancerRennesFrance
| | - Alexander Birbrair
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Dermatology, University of Wisconsin-MadisonMadisonUnited States
| | - Thierry Lamy
- Service d’hématologie Clinique, Centre Hospitalier Universitaire de RennesRennesFrance
- UMR U1236, INSERM, Université Rennes, EFS Bretagne, Equipe Labellisée Ligue Contre le CancerRennesFrance
| | - Karin Tarte
- UMR U1236, INSERM, Université Rennes, EFS Bretagne, Equipe Labellisée Ligue Contre le CancerRennesFrance
- Laboratoire Suivi Immunologique des Thérapeutiques Innovantes, Centre Hospitalier Universitaire de RennesRennesFrance
| | - Paul Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Medical Oncology, Albert Einstein College of MedicineBronxUnited States
| | - Kira Gritsman
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price CenterBronxUnited States
- Department of Medical Oncology, Albert Einstein College of MedicineBronxUnited States
| |
Collapse
|
5
|
Thompson GB, Barnhouse VR, Bierman SK, Harley BA. Influence of Hypoxia on a Biomaterial Model of the Bone Marrow Perivascular Niche. Adv Healthc Mater 2025; 14:e2500858. [PMID: 40285591 PMCID: PMC12118339 DOI: 10.1002/adhm.202500858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/15/2025] [Indexed: 04/29/2025]
Abstract
Hematopoietic stem cell (HSC) fate is shaped by distinct microenvironments termed niches within the bone marrow. Quiescence, expansion, and differentiation are directly and indirectly regulated by complex combinations of cell secretomes, cell-cell interactions, mechanical signals, and metabolic factors including oxygen tension. The perivascular environment in the bone marrow has been implicated in guiding HSC fate. However, bone marrow presents an environment which is hypoxic (≈1-4% O2) relative to traditional cell culture conditions, and the study of hypoxia in vitro is complicated by the speed with which normoxic conditions during HSC isolation induce differentiation. There is a unique opportunity to use engineered models of the bone marrow to investigate the impact of defined hypoxia on HSC fate. Here, the coordinated impact of oxygen tension and the perivascular secretome upon murine hematopoietic stem and progenitor cells (HSPCs) is examined in vitro. The findings highlight the importance of mitigating oxygen shock during cell isolation in engineered marrow models. We report a shift toward the Lineage- phenotype with hypoxic culture, expansion of HSPCs in response to perivascular niche conditioned medium, and enhanced HSPC maintenance in a hydrogel model of bone marrow in hypoxic culture when oxygen shock is mitigated during isolation using cyclosporin A.
Collapse
Affiliation(s)
- Gunnar B. Thompson
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Victoria R. Barnhouse
- Department of BioengineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Sydney K. Bierman
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Brendan A.C. Harley
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
- Department of BioengineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
- Cancer Center at IllinoisUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
- Carl R. Woese Institute for Genomic BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| |
Collapse
|
6
|
Peng B, Wang Y, Zhang H. Mitonuclear Communication in Stem Cell Function. Cell Prolif 2025; 58:e13796. [PMID: 39726221 PMCID: PMC12099226 DOI: 10.1111/cpr.13796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024] Open
Abstract
Mitochondria perform multiple functions within the cell, including the production of ATP and a great deal of metabolic intermediates, while also contributing to the cellular stress response. The majority of mitochondrial proteins are encoded by nuclear genomes, highlighting the importance of mitonuclear communication for sustaining mitochondrial homeostasis and functional. As a crucial part of the intracellular signalling network, mitochondria can impact stem cell fate determinations. Considering the essential function of stem cells in tissue maintenance, regeneration and aging, it is important to understand how mitochondria influence stem cell fate. This review explores the significant roles of mitonuclear communication and mitochondrial proteostasis, highlighting their influence on stem cells. We also examine how mitonuclear interactions contribute to cellular homeostasis, stem cell therapies, and the potential for extending lifespan.
Collapse
Affiliation(s)
- Baozhou Peng
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- The Department of Histology and Embryology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Yaning Wang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- The Department of Histology and Embryology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Hongbo Zhang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- The Department of Histology and Embryology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
7
|
Akuma M, Kim M, Zhu C, Wiljer E, Gaudreau-Lapierre A, Patterson LD, Egevad L, Tanguay S, Trinkle-Mulcahy L, Stanford WL, Riazalhosseini Y, Russell RC. Loss of VHL-mediated pRb regulation promotes clear cell renal cell carcinoma. Cell Death Dis 2025; 16:307. [PMID: 40240354 PMCID: PMC12003641 DOI: 10.1038/s41419-025-07623-y] [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: 04/14/2024] [Revised: 02/26/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Abstract
The von Hippel-Lindau (VHL) tumor suppressor is a substrate-defining component of E3 ubiquitin ligase complexes that target cellular substrates for proteasome-mediated degradation. VHL inactivation by mutation or transcriptional silencing is observed in most sporadic cases of clear cell renal cell carcinoma (ccRCC). VHL loss in ccRCC leads to constitutive stabilization of E3 ligase substrates, including hypoxia inducible factor α (HIFα). HIFα stabilization upon VHL loss is known to contribute to ccRCC development through transactivation of hypoxia-responsive genes. HIF-independent VHL targets have been implicated in oncogenesis, although those mechanisms are less well-defined than for HIFα. Using proximity labeling to identify proteasomal-sensitive VHL interactors, we identified retinoblastoma protein (pRb) as a novel substrate of VHL. Mechanistically, VHL interacts with pRb in a proteasomal-sensitive manner, promoting its ubiquitin-mediated degradation. Concordantly, VHL-inactivation results in pRb hyperstabilization. Functionally, loss of pRb in ccRCC led to increased cell death, transcriptional changes, and loss of oncogenic properties in vitro and in vivo. We also show that downstream transcriptional changes induced by pRb hyperstabilization may contribute to ccRCC tumor development. Together, our findings reveal a novel VHL-related pathway which can be therapeutically targeted to inhibit ccRCC tumor development.
Collapse
Affiliation(s)
- Mercy Akuma
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Minjun Kim
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0G1, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | - Chenxuan Zhu
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Ellis Wiljer
- Ottawa Hospital Research Institute (OHRI), Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Antoine Gaudreau-Lapierre
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Leshan D Patterson
- Department of Science, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Lars Egevad
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Simon Tanguay
- Department of Surgery, Division of Urology, McGill University, Montreal, QC, Canada
| | - Laura Trinkle-Mulcahy
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - William L Stanford
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
- Ottawa Hospital Research Institute (OHRI), Ottawa, ON, K1H 8L6, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Yasser Riazalhosseini
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0G1, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | - Ryan C Russell
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada.
- University of Ottawa Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada.
| |
Collapse
|
8
|
Zhang B, Fagarasan S. Metabolism and metabolites regulating hematopoiesis. Curr Opin Immunol 2025; 93:102525. [PMID: 39827832 DOI: 10.1016/j.coi.2025.102525] [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: 08/12/2024] [Revised: 12/24/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025]
Abstract
Energy metabolism of immune cells, such as glycolysis and mitochondrial activity, requires strict regulation. This is especially critical in the complex environment of the bone marrow (BM), where there is a need to both preserve the quiescence of hematopoietic stem cells (HSCs) and guarantee timed and effective lineage differentiation of the HSCs. Recent advances highlight the critical roles played by bioactive metabolites in regulating hematopoiesis. In particular, secreted immune metabolites (SIMets), such as γ-aminobutyric acid (GABA) and acetylcholine, secreted by B-lineage cells, act as potent modulators of hematopoietic processes, influencing HSC differentiation and emergency hematopoiesis. In this review, we provide an overview and discuss mechanisms by which energy metabolism and SIMets regulate hematopoiesis. We propose that biochemical communication facilitated by these metabolites is essential for maintaining the BM niche and suggest potential therapeutic strategies using SIMets in hematological disorders.
Collapse
Affiliation(s)
- Baihao Zhang
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan.
| | - Sidonia Fagarasan
- Laboratory for Mucosal Immunity, Center for Integrative Medical Sciences, RIKEN Yokohama Institute, Yokohama, Japan; Division of Integrated High-Order Regulatory Systems, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
9
|
Mirchandani AS, Sanchez-Garcia MA, Walmsley SR. How oxygenation shapes immune responses: emerging roles for physioxia and pathological hypoxia. Nat Rev Immunol 2025; 25:161-177. [PMID: 39349943 DOI: 10.1038/s41577-024-01087-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 03/04/2025]
Abstract
Most eukaryotes require oxygen for their survival and, with increasing multicellular complexity, oxygen availability and delivery rates vary across the tissues of complex organisms. In humans, healthy tissues have markedly different oxygen gradients, ranging from the hypoxic environment of the bone marrow (where our haematopoietic stem cells reside) to the lungs and their alveoli, which are among the most oxygenated areas of the body. Immune cells are therefore required to adapt to varying oxygen availability as they move from the bone marrow to peripheral organs to mediate their effector functions. These changing oxygen gradients are exaggerated during inflammation, where oxygenation is often depleted owing to alterations in tissue perfusion and increased cellular activity. As such, it is important to consider the effects of oxygenation on shaping the immune response during tissue homeostasis and disease conditions. In this Review, we address the relevance of both physiological oxygenation (physioxia) and disease-associated hypoxia (where cellular oxygen demand outstrips supply) for immune cell functions, discussing the relevance of hypoxia for immune responses in the settings of tissue homeostasis, inflammation, infection, cancer and disease immunotherapy.
Collapse
Affiliation(s)
- Ananda Shanti Mirchandani
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| | | | - Sarah Ruth Walmsley
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
10
|
Thompson GB, Barnhouse VR, Bierman SK, Harley BAC. Influence of Hypoxia on a Biomaterial Model of the Bone Marrow Perivascular Niche. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639296. [PMID: 40060591 PMCID: PMC11888168 DOI: 10.1101/2025.02.20.639296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Hematopoietic stem cell (HSC) fate is shaped by distinct microenvironments termed niches within the bone marrow. Quiescence, expansion, and differentiation are directly and indirectly regulated by complex combinations of cell secretomes, cell-cell interactions, mechanical signals, and metabolic factors including oxygen tension. The perivascular environment in the bone marrow has been implicated in guiding HSC fate. However, bone marrow presents an environment which is hypoxic (~1-4% O2) relative to traditional cell culture conditions, and the study of hypoxia in vitro is complicated by the speed with which normoxic conditions during HSC isolation induce differentiation. There is a unique opportunity to use engineered models of the bone marrow to investigate the impact of defined hypoxia on HSC fate. Here, we examine the coordinated impact of oxygen tension and the perivascular secretome upon murine hematopoietic stem and progenitor cells (HSPCs) in vitro. Our findings highlight the importance of mitigating oxygen shock during cell isolation in engineered marrow models. We report a shift toward the Lineage- phenotype with hypoxic culture, expansion of HSPCs in response to perivascular niche conditioned medium, and enhanced HSPC maintenance in a hydrogel model of bone marrow in hypoxic culture when oxygen shock is mitigated during isolation using cyclosporin A.
Collapse
Affiliation(s)
- Gunnar B Thompson
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Victoria R Barnhouse
- Dept. Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Sydney K Bierman
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Dept. Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
11
|
Zhao Y, Wang C, Du J, Wang W, Wu J, Liu T, Xue P, Ju Y, Hong X, Zheng J, Qu W, Zhang Y. Cadmium biphasically impacts the adaptive immune system via regulating mitochondrial activation of hematopoietic stem cells in mice. Toxicol Appl Pharmacol 2025; 495:117216. [PMID: 39725238 DOI: 10.1016/j.taap.2024.117216] [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: 10/17/2024] [Revised: 12/13/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
Cadmium (Cd) is a highly toxic metal in human body, and therefore understanding the immunotoxicity of Cd is significant for public health. The aim of this study was to investigate the role of hematopoietic stem cells (HSC) in regulating the immunotoxicity of Cd. After exposure to 10 ppm Cd via drinking water for up to 9 months, C57BL/6 mice had a suppressed adaptive immune system at day 135 but had an enhanced adaptive immune system at day 270 during Cd exposure. The biphasic impacts of Cd on the adaptive immune system were correlated to the mitochondrial (MT) activation of HSC. Mechanistically, a direct action of Cd activated the non-canonical Wnt signaling to increase MT activation in HSC in the bone marrow (BM) at day 90, thus resulting in an impaired adaptive immune system in mice at day 135 during Cd exposure; conversely, Cd reduced the production of thrombopoietin (TPO) by osteoblasts in the BM to suppress MT activation in HSC at day 180, which in turn caused an enhanced adaptive immune system in mice at day 270 during Cd exposure. Thus, Cd biphasically impacts the adaptive immune system via regulating MT activation of HSC, providing a novel angle for understanding the immunotoxicology of metals.
Collapse
Affiliation(s)
- Yifan Zhao
- Experimental Center for Research, School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Chuanxuan Wang
- Experimental Center for Research, School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Jun Du
- Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai 201203, China
| | - Wei Wang
- Experimental Center for Research, School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Jiaojiao Wu
- Experimental Center for Research, School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Ting Liu
- Experimental Center for Research, School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Peng Xue
- Experimental Center for Research, School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Yingzi Ju
- Experimental Center for Research, School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Xinyu Hong
- Institute of Chemical Toxicity Testing/State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, Shanghai Municipal Center for Disease Control and Prevention, Shanghai 200336, China.
| | - Jianheng Zheng
- Amway (Shanghai) Innovation & Science Co., Ltd, Shanghai 201203, China.
| | - Weidong Qu
- Experimental Center for Research, School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Yubin Zhang
- Experimental Center for Research, School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China.
| |
Collapse
|
12
|
Lee JH. ATM in immunobiology: From lymphocyte development to cancer immunotherapy. Transl Oncol 2025; 52:102268. [PMID: 39752906 PMCID: PMC11754496 DOI: 10.1016/j.tranon.2024.102268] [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: 08/13/2024] [Revised: 11/14/2024] [Accepted: 12/30/2024] [Indexed: 01/25/2025] Open
Abstract
Ataxia Telangiectasia Mutated (ATM) is a protein kinase traditionally known for its role in DNA damage response and cell cycle regulation. However, emerging research has revealed its multifaceted and crucial functions in the immune system. This comprehensive review explores the diverse roles of ATM in immune regulation, from lymphocyte development to its involvement in cancer immunotherapy. The review describes ATM's critical functions in V(D)J recombination and class switch recombination, highlighting its importance in adaptive immunity. It examines ATM's role in innate immunity, particularly in NF-κB signaling and cytokine production. Furthermore, the review analyzes the impact of ATM deficiency on oxidative stress and mitochondrial function in immune cells, providing insights into the immunological defects observed in Ataxia Telangiectasia (A-T). The article explores ATM's significance in maintaining hematopoietic stem cell function and its implications for bone marrow transplantation and gene therapy. Additionally, it addresses ATM's involvement in inflammation and immune senescence, linking DNA damage response to age-related immune decline. Finally, this review highlights the emerging role of ATM in cancer immunotherapy, where its inhibition shows promise in enhancing immune checkpoint blockade therapy. This review synthesizes current knowledge on ATM's functions in the immune system, offering insights into the pathophysiology of ATM-related disorders and potential therapeutic strategies for immune-related conditions and cancer immunotherapy.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, Gwangju 61186, South Korea.
| |
Collapse
|
13
|
Marchand T, Akinnola KE, Takeishi S, Maryanovich M, Pinho S, Saint-Vanne J, Birbrair A, Lamy T, Tarte K, Frenette PS, Gritsman K. Periosteal skeletal stem cells can migrate into the bone marrow and support hematopoiesis after injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.01.12.523842. [PMID: 36711927 PMCID: PMC9882153 DOI: 10.1101/2023.01.12.523842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Skeletal stem cells have been isolated from various tissues, including periosteum and bone marrow, where they exhibit key functions in bone biology and hematopoiesis, respectively. The role of periosteal skeletal stem cells in bone regeneration and healing has been extensively studied, but their ability to contribute to the bone marrow stroma is still under debate. In the present study, we characterized a whole bone transplantation model that mimics the initial bone marrow necrosis and fatty infiltration seen after injury. Using this model and a lineage tracing approach, we observed the migration of periosteal skeletal stem cells into the bone marrow after transplantation. Once in the bone marrow, periosteal skeletal stem cells are phenotypically and functionally reprogrammed into bone marrow mesenchymal stem cells that express high levels of hematopoietic stem cell niche factors such as Cxcl12 and Kitl. In addition, using ex vivo and in vivo approaches, we found that periosteal skeletal stem cells are more resistant to acute stress than bone marrow mesenchymal stem cells. These results highlight the plasticity of periosteal skeletal stem cells and their potential role in bone marrow regeneration after bone marrow injury.
Collapse
Affiliation(s)
- Tony Marchand
- Service d’hématologie Clinique, Centre Hospitalier Universitaire de Rennes, Rennes, France
- UMR U1236, INSERM, Université Rennes, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price Center, 1301 Morris Park Avenue, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price Center, 1300 Morris Park Avenue, Room 101, Bronx, NY 10461, USA
| | - Kemi E. Akinnola
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price Center, 1301 Morris Park Avenue, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price Center, 1300 Morris Park Avenue, Room 101, Bronx, NY 10461, USA
| | - Shoichiro Takeishi
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price Center, 1301 Morris Park Avenue, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price Center, 1300 Morris Park Avenue, Room 101, Bronx, NY 10461, USA
| | - Maria Maryanovich
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price Center, 1301 Morris Park Avenue, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price Center, 1300 Morris Park Avenue, Room 101, Bronx, NY 10461, USA
| | - Sandra Pinho
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price Center, 1301 Morris Park Avenue, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price Center, 1300 Morris Park Avenue, Room 101, Bronx, NY 10461, USA
- Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pharmacology & Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Julien Saint-Vanne
- UMR U1236, INSERM, Université Rennes, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
| | - Alexander Birbrair
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price Center, 1301 Morris Park Avenue, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price Center, 1300 Morris Park Avenue, Room 101, Bronx, NY 10461, USA
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Thierry Lamy
- Service d’hématologie Clinique, Centre Hospitalier Universitaire de Rennes, Rennes, France
- UMR U1236, INSERM, Université Rennes, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
| | - Karin Tarte
- UMR U1236, INSERM, Université Rennes, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
- Laboratoire Suivi Immunologique des Thérapeutiques Innovantes, Centre Hospitalier Universitaire de Rennes, F-35033 Rennes, France
| | - Paul S. Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price Center, 1301 Morris Park Avenue, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price Center, 1300 Morris Park Avenue, Room 101, Bronx, NY 10461, USA
- Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kira Gritsman
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Michael F. Price Center, 1301 Morris Park Avenue, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Michael F. Price Center, 1300 Morris Park Avenue, Room 101, Bronx, NY 10461, USA
- Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
14
|
Li J, Bai J, Pham VT, Hashimoto M, Sezaki M, Shi Q, Jin Q, He C, Armstrong A, Li T, Pan M, Liu S, Luan Y, Zeng H, Andreassen PR, Huang G. Loss of Cpt1a results in elevated glucose-fueled mitochondrial oxidative phosphorylation and defective hematopoietic stem cells. J Clin Invest 2025; 135:e184069. [PMID: 39786963 PMCID: PMC11870731 DOI: 10.1172/jci184069] [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: 06/21/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
Hematopoietic stem cells (HSCs) rely on self-renewal to sustain stem cell potential and undergo differentiation to generate mature blood cells. Mitochondrial fatty acid β-oxidation (FAO) is essential for HSC maintenance. However, the role of carnitine palmitoyl transferase 1a (CPT1A), a key enzyme in FAO, remains unclear in HSCs. Using a Cpt1a hematopoiesis-specific conditional-KO (Cpt1aΔ/Δ) mouse model, we found that loss of Cpt1a led to HSC defects, including loss of HSC quiescence and self-renewal and increased differentiation. Mechanistically, we found that loss of Cpt1a resulted in elevated levels of mitochondrial respiratory chain complex components and their activity, as well as increased ATP production and accumulation of mitochondrial ROS in HSCs. Taken together, this suggests hyperactivation of mitochondria and metabolic rewiring via upregulated glucose-fueled oxidative phosphorylation (OXPHOS). In summary, our findings demonstrate an essential role for Cpt1a in HSC maintenance and provide insight into the regulation of mitochondrial metabolism via control of the balance between FAO and glucose-fueled OXPHOS.
Collapse
Affiliation(s)
- Jue Li
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
| | - Jie Bai
- Department of Cell Systems and Anatomy, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
- Mays Cancer Center at UT Health San Antonio, San Antonio, Texas, USA
| | - Vincent T. Pham
- Department of Cell Systems and Anatomy, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
- Mays Cancer Center at UT Health San Antonio, San Antonio, Texas, USA
| | - Michihiro Hashimoto
- Department of Cell Systems and Anatomy, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
- Mays Cancer Center at UT Health San Antonio, San Antonio, Texas, USA
- Department of Pathology and Laboratory Medicine, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
| | - Maiko Sezaki
- Department of Cell Systems and Anatomy, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
- Mays Cancer Center at UT Health San Antonio, San Antonio, Texas, USA
| | - Qili Shi
- Department of Cell Systems and Anatomy, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
| | - Qiushi Jin
- Department of Cell Systems and Anatomy, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
| | - Chenhui He
- Department of Cell Systems and Anatomy, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
| | - Amy Armstrong
- Department of Cell Systems and Anatomy, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
- Department of Microbiology, Immunology and Molecular Genetics, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
| | - Tian Li
- Department of Cell Systems and Anatomy, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
| | - Mingzhe Pan
- Department of Cell Systems and Anatomy, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
| | - Shujun Liu
- Department of Medicine, The MetroHealth System, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yu Luan
- Department of Cell Systems and Anatomy, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
- Mays Cancer Center at UT Health San Antonio, San Antonio, Texas, USA
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Hui Zeng
- Department of Hematology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Paul R. Andreassen
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Gang Huang
- Department of Cell Systems and Anatomy, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
- Mays Cancer Center at UT Health San Antonio, San Antonio, Texas, USA
- Department of Pathology and Laboratory Medicine, UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, Texas, USA
| |
Collapse
|
15
|
Joly A, Schott A, Phadke I, Gonzalez-Menendez P, Kinet S, Taylor N. Beyond ATP: Metabolite Networks as Regulators of Physiological and Pathological Erythroid Differentiation. Physiology (Bethesda) 2025; 40:0. [PMID: 39226028 DOI: 10.1152/physiol.00035.2024] [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: 07/15/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
Hematopoietic stem cells (HSCs) possess the capacity for self-renewal and the sustained production of all mature blood cell lineages. It has been well established that a metabolic rewiring controls the switch of HSCs from a self-renewal state to a more differentiated state, but it is only recently that we have appreciated the importance of metabolic pathways in regulating the commitment of progenitors to distinct hematopoietic lineages. In the context of erythroid differentiation, an extensive network of metabolites, including amino acids, sugars, nucleotides, fatty acids, vitamins, and iron, is required for red blood cell (RBC) maturation. In this review, we highlight the multifaceted roles via which metabolites regulate physiological erythropoiesis as well as the effects of metabolic perturbations on erythroid lineage commitment and differentiation. Of note, the erythroid differentiation process is associated with an exceptional breadth of solute carrier (SLC) metabolite transporter upregulation. Finally, we discuss how recent research, revealing the critical impact of metabolic reprogramming in diseases of disordered and ineffective erythropoiesis, has created opportunities for the development of novel metabolic-centered therapeutic strategies.
Collapse
Affiliation(s)
- Axel Joly
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Arthur Schott
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Ira Phadke
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Pediatric Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Pedro Gonzalez-Menendez
- Departamento de Morfologia y Biologia Celular, Instituto Universitario de Oncologia del Principado de Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Sandrina Kinet
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Naomi Taylor
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Pediatric Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
16
|
Kazerani M, Cernilogar F, Pasquarella A, Hinterberger M, Nuber A, Klein L, Schotta G. Histone methyltransferase SETDB1 safeguards mouse fetal hematopoiesis by suppressing activation of cryptic enhancers. Proc Natl Acad Sci U S A 2024; 121:e2409656121. [PMID: 39689172 DOI: 10.1073/pnas.2409656121] [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: 05/14/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
The H3K9me3-specific histone methyltransferase SETDB1 is critical for proper regulation of developmental processes, but the underlying mechanisms are only partially understood. Here, we show that deletion of Setdb1 in mouse fetal liver hematopoietic stem and progenitor cells (HSPCs) results in compromised stem cell function, enhanced myeloerythroid differentiation, and impaired lymphoid development. Notably, Setdb1-deficient HSPCs exhibit reduced quiescence and increased proliferation, accompanied by the acquisition of a lineage-biased transcriptional program. In Setdb1-deficient HSPCs, we identify genomic regions that are characterized by loss of H3K9me3 and increased chromatin accessibility, suggesting enhanced transcription factor (TF) activity. Interestingly, hematopoietic TFs like PU.1 bind these cryptic enhancers in wild-type HSPCs, despite the H3K9me3 status. Thus, our data indicate that SETDB1 restricts activation of nonphysiological TF binding sites which helps to ensure proper maintenance and differentiation of fetal liver HSPCs.
Collapse
Affiliation(s)
- Maryam Kazerani
- Division of Molecular Biology, Biomedical Center, Faculty of Medicine, Ludwig Maximilian University Munich, Martinsried 82152, Germany
| | - Filippo Cernilogar
- Division of Molecular Biology, Biomedical Center, Faculty of Medicine, Ludwig Maximilian University Munich, Martinsried 82152, Germany
- Department of Science and Technological Innovation, University of Piemonte Orientale, Alessandria 15121, Italy
| | - Alessandra Pasquarella
- Division of Molecular Biology, Biomedical Center, Faculty of Medicine, Ludwig Maximilian University Munich, Martinsried 82152, Germany
| | - Maria Hinterberger
- Institute for Immunology, Biomedical Center, Faculty of Medicine, Ludwig Maximilian University Munich, Martinsried 82152, Germany
| | - Alexander Nuber
- Division of Molecular Biology, Biomedical Center, Faculty of Medicine, Ludwig Maximilian University Munich, Martinsried 82152, Germany
| | - Ludger Klein
- Institute for Immunology, Biomedical Center, Faculty of Medicine, Ludwig Maximilian University Munich, Martinsried 82152, Germany
| | - Gunnar Schotta
- Division of Molecular Biology, Biomedical Center, Faculty of Medicine, Ludwig Maximilian University Munich, Martinsried 82152, Germany
| |
Collapse
|
17
|
Liu X, Liu Y, Rao Q, Mei Y, Xing H, Gu R, Mou J, Chen M, Ding F, Xie W, Tang K, Tian Z, Wang M, Qiu S, Wang J. Targeting Fatty Acid Metabolism Abrogates the Differentiation Blockade in Preleukemic Cells. Cancer Res 2024; 84:4233-4245. [PMID: 39264725 DOI: 10.1158/0008-5472.can-23-3861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/29/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024]
Abstract
Metabolism plays a key role in the maintenance of normal hematopoietic stem cells (HSC) and in the development of leukemia. A better understanding of the metabolic characteristics and dependencies of preleukemic cells could help identify potential therapeutic targets to prevent leukemic transformation. As AML1-ETO, one of the most frequent fusion proteins in acute myeloid leukemia that is encoded by a RUNX1::RUNX1T1 fusion gene, is capable of generating preleukemic clones, in this study, we used a conditional Runx1::Runx1t1 knockin mouse model to evaluate preleukemic cell metabolism. AML1-ETO expression resulted in impaired hematopoietic reconstitution and increased self-renewal ability. Oxidative phosphorylation and glycolysis decreased significantly in these preleukemic cells accompanied by increased HSC quiescence and reduced cell cycling. Furthermore, HSCs expressing AML1-ETO exhibited an increased requirement for fatty acids through metabolic flux. Dietary lipid deprivation or loss of the fatty acid transporter FATP3 by targeted deletion using CRISPR/Cas9 partially restored differentiation. These findings reveal the unique metabolic profile of preleukemic cells and propose FATP3 as a potential target for disrupting leukemogenesis. Significance: Fatty acid metabolism is required for maintenance of preleukemic cells but dispensable for normal hematopoiesis, indicating that dietary lipid deprivation or inhibiting fatty acid uptake may serve as potential strategies to prevent leukemogenesis.
Collapse
Affiliation(s)
- Xiaoyu Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yu Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Qing Rao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yihan Mei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Runxia Gu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Junli Mou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Manling Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Fan Ding
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wanqing Xie
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Kejing Tang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Zheng Tian
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Min Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shaowei Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
18
|
Thorsell A, Sjölin L, Berger E, Jeppsson A, Oldfors A, Rotter Sopasakis V, Vukusic K. Stem Cell-Associated Proteins and Extracellular Matrix Composition of the Human Atrioventricular Junction. Cells 2024; 13:2048. [PMID: 39768140 PMCID: PMC11674807 DOI: 10.3390/cells13242048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The human heart regenerates slowly through life, but how new cells are generated is mostly unknown. The atrioventricular junction (AVj) has been indicated as a potential stem cell niche region. Little is known about the protein composition of the human AVj. To map the extracellular matrix (ECM) and expression of stem cell-related biomarkers, this study compares protein and gene expression patterns in AVj and Left Ventricular (LV) tissues. Biopsies were collected from 15 human hearts. Global quantitative proteomics and mRNA sequencing were used to identify differentially expressed proteins and altered genes. Of the total 4904 identified proteins, 1138 were differently expressed between the AVj and LV. While the top proteins in LV were involved in cardiac motor function and energy regulation, the AVj displayed proteins associated with early cardiomyocyte development, differentiation, proliferation, migration, and hypoxia. Furthermore, several developmental signalling pathways, including TGF-β, TNF, WNT, Notch, and FGF, were represented. RNA-seq data verified that the expressed genes were involved with differentiation, cell growth, proliferation, or ECM organization. Immunohistochemistry confirmed the expression of the stem cell-related biomarkers NPPA and POSTN in the AVj, further strengthening the hypothesis of the AVj as a specialized microenvironment conducive to stem cell niche activity.
Collapse
Affiliation(s)
- Annika Thorsell
- Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Linnéa Sjölin
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Evelin Berger
- Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Anders Jeppsson
- Region Västra Götaland, Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Anders Oldfors
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden
- Department of Pathology, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Victoria Rotter Sopasakis
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden
- Region Västra Götaland, Department of Clinical Chemistry, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Kristina Vukusic
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden
| |
Collapse
|
19
|
Ruffinatto L, Groult Y, Iacono J, Sarrazin S, de Laval B. Hematopoietic stem cell a reservoir of innate immune memory. Front Immunol 2024; 15:1491729. [PMID: 39720722 PMCID: PMC11666435 DOI: 10.3389/fimmu.2024.1491729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/30/2024] [Indexed: 12/26/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are a rare, long-lived and multipotent population that give rise to majority of blood cells and some tissue-resident immune cells. There is growing evidence that inflammatory stimuli can trigger persistent reprogramming in HSCs that enhances or inhibits the cellular functions of these HSCs and their progeny in response to subsequent infections. This newly discovered property makes HSCs a reservoir for innate immune memory. The molecular mechanisms underlying innate immune memory in HSCs are similar to those observed in innate immune cells, although their full elucidation is still pending. In this review, we examine the current state of knowledge on how an inflammatory response leads to reprogramming of HSCs. Understanding the full spectrum of consequences of reshaping early hematopoiesis is critical for assessing the potential benefits and risks under physiological and pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | - Bérengère de Laval
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut
National de la Santé et de la Recherche Médicale (INSERM), Centre d’Immunologie de Marseille-Luminy (CIML), Marseille, France
| |
Collapse
|
20
|
Zhou H, Xiang W, Zhou G, Rodrigues-Lima F, Guidez F, Wang L. Metabolic dysregulation in myelodysplastic neoplasm: impact on pathogenesis and potential therapeutic targets. Med Oncol 2024; 42:23. [PMID: 39644425 DOI: 10.1007/s12032-024-02575-3] [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: 10/09/2024] [Accepted: 11/23/2024] [Indexed: 12/09/2024]
Abstract
Despite significant advancements in the research of the pathogenesis mechanisms of Myelodysplastic Neoplasm (MDS) in recent years, there are still many gaps to fill. The advancement of metabolomics studies has led to a research booming in clarifying the impact of metabolic abnormalities during the pathogenesis of MDS. The present review primarily focuses on the dysregulated metabolic pathways, exploring the influences on the pathogenesis of MDS and their roles during the course of the disease. Furthermore, we discuss the potential of relevant metabolic pathways as therapeutic targets, along with the latest metabolic-related treatment drugs and approaches.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Wenqiong Xiang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Guangyu Zhou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Fernando Rodrigues-Lima
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle Et Adaptative, 75013, Paris, France
| | - Fabien Guidez
- Université Paris Cité, Institut de Recherche Saint Louis INSERM UMR_S1131, 75010, Paris, France
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
21
|
Herrera J, Bensussen A, García-Gómez ML, Garay-Arroyo A, Álvarez-Buylla ER. A system-level model reveals that transcriptional stochasticity is required for hematopoietic stem cell differentiation. NPJ Syst Biol Appl 2024; 10:145. [PMID: 39639033 PMCID: PMC11621455 DOI: 10.1038/s41540-024-00469-8] [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: 05/09/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
Abstract
HSCs differentiation has been difficult to study experimentally due to the high number of components and interactions involved, as well as the impact of diverse physiological conditions. From a 200-node network, that was grounded on experimental data, we derived a 21-node regulatory network by collapsing linear pathways and retaining the functional feedback loops. This regulatory network core integrates key nodes and interactions underlying HSCs differentiation, including transcription factors, metabolic, and redox signaling pathways. We used Boolean, continuous, and stochastic dynamic models to simulate the hypoxic conditions of the HSCs niche, as well as the patterns and temporal sequences of HSCs transitions and differentiation. Our findings indicate that HSCs differentiation is a plastic process in which cell fates can transdifferentiate among themselves. Additionally, we found that cell heterogeneity is fundamental for HSCs differentiation. Lastly, we found that oxygen activates ROS production, inhibiting quiescence and promoting growth and differentiation pathways of HSCs.
Collapse
Affiliation(s)
- Joel Herrera
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Antonio Bensussen
- Departamento de Control Automático, Cinvestav-IPN, Ciudad de México, México
| | - Mónica L García-Gómez
- Theoretical Biology, Institute of Biodynamics and Biocomplexity; Experimental and Computational Plant Development, Institute of Environmental Biology, Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Adriana Garay-Arroyo
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Elena R Álvarez-Buylla
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, México.
| |
Collapse
|
22
|
Rodriguez-Sevilla JJ, Colla S. Inflammation in myelodysplastic syndrome pathogenesis. Semin Hematol 2024; 61:385-396. [PMID: 39424469 DOI: 10.1053/j.seminhematol.2024.09.005] [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: 09/07/2024] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
Inflammation is a key driver of the progression of preleukemic myeloid conditions, such as clonal hematopoiesis of indeterminate potential (CHIP) and clonal cytopenia of undetermined significance (CCUS), to myelodysplastic syndromes (MDS). Inflammation is a critical mediator in the complex interplay of the genetic, epigenetic, and microenvironmental factors contributing to clonal evolution. Under inflammatory conditions, somatic mutations in TET2, DNMT3A, and ASXL1, the most frequently mutated genes in CHIP and CCUS, induce a competitive advantage to hematopoietic stem and progenitor cells, which leads to their clonal expansion in the bone marrow. Chronic inflammation also drives metabolic reprogramming and immune system deregulation, further promoting the expansion of malignant clones. This review underscores the urgent need to fully elucidate the role of inflammation in MDS initiation and highlights the potential of the therapeutical targeting of inflammatory pathways as an early intervention in MDS.
Collapse
Affiliation(s)
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
23
|
Cui Y, Ren Y, Ren F, Zhang Y, Wang H. Synergistic effect and molecular mechanism of nicotinamide and UM171 in ex vivo expansion of long-term hematopoietic stem cells. Regen Ther 2024; 27:191-199. [PMID: 38840730 PMCID: PMC11150914 DOI: 10.1016/j.reth.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/10/2024] [Accepted: 03/15/2024] [Indexed: 06/07/2024] Open
Abstract
Introduction Several approaches to expand human hematopoietic stem cells (HSCs) have been reported, but the ability of these methods to expand long-term hematopoietic stem cells (LT-HSCs) remains to be improved, which limits the application of HSCs-based therapies. Methods CD34+ cells were purified from umbilical cord blood using MacsCD34 beads, and then cultured for 12 d in a serum-free medium. Flow cytometry was used to detect phenotype, cell cycle distribution, and apoptosis of the cultured cells. Colony-forming cell (CFC) assays can evaluate multi-lineage differentiation potential of HSCs. Real-time polymerase chain reaction was employed to detect the expression of genes related to self-renewal programs and antioxidant activity. DCFH-DA probes were used to evaluate intracellular production of reactive oxygen species (ROS). Determination of the effect of different culture conditions on the balance of cytokine by cytometric bead array. Results Here, we show a combination, Nicotinamide (NAM) combined with pyrimidoindole derivative UM171, can massively expand LT-HSCs ex vivo, and the expanded cells maintained the capability of self-renewal and multilineage differentiation. Additionally, our data indicated that UM171 promoted self-renewal of HSCs by inducing HSCs entry into the cell cycle and activating Notch and Wnt pathways, but the infinite occurrence of this process may lead to mitochondrial metabolism disorder and differentiation of HSCs. NAM kept HSCs in their primitive and dormant states by reducing intracellular ROS levels and upregulating the expression of stemness related genes, so we believed that NAM can act as a brake to control the above process. Conclusions The discovery of the synergistic effect of NAM and UM171 for expanding LT-HSCs provides a new strategy in solving the clinical issue of limited numbers of HSCs.
Collapse
Affiliation(s)
- Yanni Cui
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yan Ren
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Fanggang Ren
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Key Laboratory of Molecular Diagnosis and Treatment of Blood Diseases in Shanxi Province, Taiyuan, China
| | - Yaofang Zhang
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Key Laboratory of Molecular Diagnosis and Treatment of Blood Diseases in Shanxi Province, Taiyuan, China
| | - Hongwei Wang
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
- Key Laboratory of Molecular Diagnosis and Treatment of Blood Diseases in Shanxi Province, Taiyuan, China
- Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
24
|
Deng J, Tan Y, Xu Z, Wang H. Advances in hematopoietic stem cells ex vivo expansion associated with bone marrow niche. Ann Hematol 2024; 103:5035-5057. [PMID: 38684510 DOI: 10.1007/s00277-024-05773-1] [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: 10/08/2023] [Accepted: 04/19/2024] [Indexed: 05/02/2024]
Abstract
Hematopoietic stem cells (HSCs) are an ideal source for the treatment of many hematological diseases and malignancies, as well as diseases of other systems, because of their two important features, self-renewal and multipotential differentiation, which have the ability to rebuild the blood system and immune system of the body. However, so far, the insufficient number of available HSCs, whether from bone marrow (BM), mobilized peripheral blood or umbilical cord blood, is still the main restricting factor for the clinical application. Therefore, strategies to expand HSCs numbers and maintain HSCs functions through ex vivo culture are urgently required. In this review, we outline the basic biology characteristics of HSCs, and focus on the regulatory factors in BM niche affecting the functions of HSCs. Then, we introduce several representative strategies used for HSCs from these three sources ex vivo expansion associated with BM niche. These findings have deepened our understanding of the mechanisms by which HSCs balance self-renewal and differentiation and provided a theoretical basis for the efficient clinical HSCs expansion.
Collapse
Affiliation(s)
- Ju Deng
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhong Tan
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhifang Xu
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hongwei Wang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
25
|
Yu L, Wei W, Lv J, Lu Y, Wang Z, Cai C. FABP4-mediated lipid metabolism promotes TNBC progression and breast cancer stem cell activity. Cancer Lett 2024; 604:217271. [PMID: 39306229 DOI: 10.1016/j.canlet.2024.217271] [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: 04/19/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Metabolic remodeling is a pivotal feature of cancer, with cancer stem cells frequently showcasing distinctive metabolic behaviors. Nonetheless, understanding the metabolic intricacies of triple-negative breast cancer (TNBC) and breast cancer stem cells (BCSCs) has remained elusive. In this study, we meticulously characterized the metabolic profiles of TNBC and BCSCs and delved into their potential implications for TNBC treatment. Our findings illuminated the robust lipid metabolism activity within TNBC tumors, especially in BCSCs. Furthermore, we discovered that Fabp4, through its mediation of fatty acid uptake, plays a crucial role in regulating TNBC lipid metabolism. Knocking down Fabp4 or inhibiting its activity significantly suppressed TNBC tumor progression in both the MMTV-Wnt1 spontaneous TNBC model and the TNBC patient-derived xenograft model. Mechanistically, Fabp4's influence on TNBC tumor progression was linked to its regulation of mitochondrial stability, the CPT1-mediated fatty acid oxidation process, and ROS production. Notably, in a high-fat diet model, Fabp4 deficiency proved to be a substantial inhibitor of obesity-accelerated TNBC progression. Collectively, these findings shed light on the unique metabolic patterns of TNBC and BCSCs, underscore the biological significance of Fabp4-mediated fatty acid metabolism in governing TNBC progression, and offer a solid theoretical foundation for considering metabolic interventions in breast cancer treatment. SIGNIFICANCE: Triple-negative breast cancer progression and breast cancer stem cell activity can be restricted by targeting a critical regulator of lipid responses, FABP4.
Collapse
Affiliation(s)
- Liya Yu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Wei Wei
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Jian Lv
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518057, China
| | - Yu Lu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Zhihua Wang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518057, China
| | - Cheguo Cai
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
26
|
Du C, Liu C, Yu K, Zhang S, Fu Z, Chen X, Liao W, Chen J, Zhang Y, Wang X, Chen M, Chen F, Shen M, Wang C, Chen S, Wang S, Wang J. Mitochondrial serine catabolism safeguards maintenance of the hematopoietic stem cell pool in homeostasis and injury. Cell Stem Cell 2024; 31:1484-1500.e9. [PMID: 39181130 DOI: 10.1016/j.stem.2024.07.009] [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: 01/18/2024] [Revised: 06/14/2024] [Accepted: 07/30/2024] [Indexed: 08/27/2024]
Abstract
Hematopoietic stem cells (HSCs) employ a very unique metabolic pattern to maintain themselves, while the spectrum of their metabolic adaptations remains incompletely understood. Here, we uncover a distinct and heterogeneous serine metabolism within HSCs and identify mouse HSCs as a serine auxotroph whose maintenance relies on exogenous serine and the ensuing mitochondrial serine catabolism driven by the hydroxymethyltransferase 2 (SHMT2)-methylene-tetrahydrofolate dehydrogenase 2 (MTHFD2) axis. Mitochondrial serine catabolism primarily feeds NAD(P)H generation to maintain redox balance and thereby diminishes ferroptosis susceptibility of HSCs. Dietary serine deficiency, or genetic or pharmacological inhibition of the SHMT2-MTHFD2 axis, increases ferroptosis susceptibility of HSCs, leading to impaired maintenance of the HSC pool. Moreover, exogenous serine protects HSCs from irradiation-induced myelosuppressive injury by fueling mitochondrial serine catabolism to mitigate ferroptosis. These findings reframe the canonical view of serine from a nonessential amino acid to an essential niche metabolite for HSC pool maintenance.
Collapse
Affiliation(s)
- Changhong Du
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Chaonan Liu
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China; Frontier Medical Training Brigade, Army Medical University (Third Military Medical University), Xinjiang 831200, China
| | - Kuan Yu
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Shuzhen Zhang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zeyu Fu
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xinliang Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Weinian Liao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jun Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yimin Zhang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xinmiao Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China; Department of Hematology, The General Hospital of Western Theater Command, Chengdu, Sichuan 610008, China
| | - Mo Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Fang Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Mingqiang Shen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Cheng Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Shilei Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Song Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
27
|
Fateh K, Mansoori F, Atashi A. The Evaluation of Mass/DNA Copy Number of Mitochondria in Umbilical Cord Blood-derived Hematopoietic Stem Cells Cocultured with MSCs. Indian J Hematol Blood Transfus 2024; 40:638-646. [PMID: 39469179 PMCID: PMC11512953 DOI: 10.1007/s12288-024-01774-2] [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: 01/15/2024] [Accepted: 04/08/2024] [Indexed: 10/30/2024] Open
Abstract
Over recent decades, UCB has been widely used as an excellent alternative source of HSCs for treating many hematologic disorders. Recent studies suggest using mesenchymal stroma cell co-cultures to increase the number of HSCs prior to transplantation. Considering the critical role of mitochondria in the cell's fate and the importance of the self-renewal capacity of HSCs in HSCT, we decided to investigate the mass/DNA copy number of mitochondria in HSCs while co-cultured with MSCs and alone after seven days. UCB units were collected from full-term deliveries. MSCs and HSCs were isolated from UCB and the purity of cells was confirmed by flow cytometry. The mtDNA-Copy Number of HSCs was calculated using prob-based real-time PCR. Furthermore, Mito Tracker Green dye measured the mass of mitochondria of HSCs. HSCs from MSC co-culture group showed significantly fewer mtDNA-CN compared to HSCs alone after seven days (p < 0.001). Besides, by comparing the two groups on day seven to HSCs on day zero, we observed a mild increase in the mitochondrial mass of HSCs alone compared to the MSC-HSC co-culture group (p < 0.05). Concerning previous studies that have proved the association between lower mass/DNA-copy number of mitochondria in CD34 + HSCs and lower metabolic activity along with higher quiescence maintenance, and by considering the results of this experiment, it seems that the MSC-HSC co-cultures might be associated with a higher expansion of HSCs as well as stemness maintenance leading to the improvement in engraftment. Nevertheless, further investigations are required to clarify the exact connection between lower mass/DNA-copy number of mitochondria and stemness maintenance in HSCs.
Collapse
Affiliation(s)
- Kosar Fateh
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mansoori
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Atashi
- Stem Cell and Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
28
|
Song Z, Park SH, Mu WC, Feng Y, Wang CL, Wang Y, Barthez M, Maruichi A, Guo J, Yang F, Lin AW, Heydari K, Chini CCS, Chini EN, Jang C, Chen D. An NAD +-dependent metabolic checkpoint regulates hematopoietic stem cell activation and aging. NATURE AGING 2024; 4:1384-1393. [PMID: 39044033 PMCID: PMC11565225 DOI: 10.1038/s43587-024-00670-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 06/18/2024] [Indexed: 07/25/2024]
Abstract
How hematopoietic stem cells (HSCs) maintain metabolic homeostasis to support tissue repair and regeneration throughout the lifespan is elusive. Here, we show that CD38, an NAD+-dependent metabolic enzyme, promotes HSC proliferation by inducing mitochondrial Ca2+ influx and mitochondrial metabolism in young mice. Conversely, aberrant CD38 upregulation during aging is a driver of HSC deterioration in aged mice due to dysregulated NAD+ metabolism and compromised mitochondrial stress management. The mitochondrial calcium uniporter, a mediator of mitochondrial Ca2+ influx, also supports HSC proliferation in young mice yet drives HSC decline in aged mice. Pharmacological inactivation of CD38 reverses HSC aging and the pathophysiological changes of the aging hematopoietic system in aged mice. Together, our study highlights an NAD+ metabolic checkpoint that balances mitochondrial activation to support HSC proliferation and mitochondrial stress management to enhance HSC self-renewal throughout the lifespan, and links aberrant Ca2+ signaling to HSC aging.
Collapse
Affiliation(s)
- Zehan Song
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
- Metabolic Biology Graduate Program, University of California, Berkeley, CA, USA
| | - Sang Hee Park
- Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - Wei-Chieh Mu
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
- Endocrinology Graduate Program, University of California, Berkeley, CA, USA
| | - Yufan Feng
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
| | - Chih-Ling Wang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
| | - Yifei Wang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
- Metabolic Biology Graduate Program, University of California, Berkeley, CA, USA
| | - Marine Barthez
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
| | - Ayane Maruichi
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
- Endocrinology Graduate Program, University of California, Berkeley, CA, USA
| | - Jiayue Guo
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
| | - Fanghan Yang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
- Endocrinology Graduate Program, University of California, Berkeley, CA, USA
| | - Anita Wong Lin
- Cancer Research Laboratory, University of California, Berkeley, CA, USA
| | - Kartoosh Heydari
- Cancer Research Laboratory, University of California, Berkeley, CA, USA
| | - Claudia C S Chini
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Eduardo N Chini
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - Danica Chen
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA.
- Metabolic Biology Graduate Program, University of California, Berkeley, CA, USA.
- Endocrinology Graduate Program, University of California, Berkeley, CA, USA.
| |
Collapse
|
29
|
Meader E, Walcheck MT, Leder MR, Jing R, Wrighton PJ, Sugden WW, Najia MA, Oderberg IM, Taylor VM, LeBlanc ZC, Quenzer ED, Lim SE, Daley GQ, Goessling W, North TE. Bnip3lb-driven mitophagy sustains expansion of the embryonic hematopoietic stem cell pool. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614531. [PMID: 39386657 PMCID: PMC11463499 DOI: 10.1101/2024.09.23.614531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Embryonic hematopoietic stem and progenitor cells (HSPCs) have the unique ability to undergo rapid proliferation while maintaining multipotency, a clinically-valuable quality which currently cannot be replicated in vitro. Here, we show that embryonic HSPCs achieve this state by precise spatio-temporal regulation of reactive oxygen species (ROS) via Bnip3lb-associated developmentally-programmed mitophagy, a distinct autophagic regulatory mechanism from that of adult HSPCs. While ROS drives HSPC specification in the dorsal aorta, scRNAseq and live-imaging of Tg(ubi:mitoQC) zebrafish indicate that mitophagy initiates as HSPCs undergo endothelial-to-hematopoietic transition and colonize the caudal hematopoietic tissue (CHT). Knockdown of bnip3lb reduced mitophagy and HSPC numbers in the CHT by promoting myeloid-biased differentiation and apoptosis, which was rescued by anti-oxidant exposure. Conversely, induction of mitophagy enhanced both embryonic HSPC and lymphoid progenitor numbers. Significantly, mitophagy activation improved ex vivo functional capacity of hematopoietic progenitors derived from human-induced pluripotent stem cells (hiPSCs), enhancing serial-replating hematopoietic colony forming potential. HIGHLIGHTS ROS promotes HSPC formation in the dorsal aorta but negatively affects maintenance thereafter.HSPCs colonizing secondary niches control ROS levels via Bnip3lb-directed mitophagy.Mitophagy protects nascent HSPCs from ROS-associated apoptosis and maintains multipotency.Induction of mitophagy enhances long-term hematopoietic potential of iPSC-derived HSPCs.
Collapse
|
30
|
Patel KD, Keskin-Erdogan Z, Sawadkar P, Nik Sharifulden NSA, Shannon MR, Patel M, Silva LB, Patel R, Chau DYS, Knowles JC, Perriman AW, Kim HW. Oxidative stress modulating nanomaterials and their biochemical roles in nanomedicine. NANOSCALE HORIZONS 2024; 9:1630-1682. [PMID: 39018043 DOI: 10.1039/d4nh00171k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Many pathological conditions are predominantly associated with oxidative stress, arising from reactive oxygen species (ROS); therefore, the modulation of redox activities has been a key strategy to restore normal tissue functions. Current approaches involve establishing a favorable cellular redox environment through the administration of therapeutic drugs and redox-active nanomaterials (RANs). In particular, RANs not only provide a stable and reliable means of therapeutic delivery but also possess the capacity to finely tune various interconnected components, including radicals, enzymes, proteins, transcription factors, and metabolites. Here, we discuss the roles that engineered RANs play in a spectrum of pathological conditions, such as cancer, neurodegenerative diseases, infections, and inflammation. We visualize the dual functions of RANs as both generator and scavenger of ROS, emphasizing their profound impact on diverse cellular functions. The focus of this review is solely on inorganic redox-active nanomaterials (inorganic RANs). Additionally, we deliberate on the challenges associated with current RANs-based approaches and propose potential research directions for their future clinical translation.
Collapse
Affiliation(s)
- Kapil D Patel
- John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, UK
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
| | - Zalike Keskin-Erdogan
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
- Department of Chemical Engineering, Imperial College London, Exhibition Rd, South Kensington, SW7 2BX, London, UK
| | - Prasad Sawadkar
- Division of Surgery and Interventional Science, UCL, London, UK
- The Griffin Institute, Northwick Park Institute for Medical Research, Northwick Park and St Mark's Hospitals, London, HA1 3UJ, UK
| | - Nik Syahirah Aliaa Nik Sharifulden
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Mark Robert Shannon
- John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, UK
| | - Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Women University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Lady Barrios Silva
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Rajkumar Patel
- Energy & Environment Sciences and Engineering (EESE), Integrated Sciences and Engineering Division (ISED), Underwood International College, Yonsei University, 85 Songdongwahak-ro, Yeonsungu, Incheon 21938, Republic of Korea
| | - David Y S Chau
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Jonathan C Knowles
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Adam W Perriman
- John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, UK
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
31
|
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
|
32
|
Han R, Yang X, Ji X, Zhou B. Remote ischemic preconditioning prevents high-altitude cerebral edema by enhancing glucose metabolic reprogramming. CNS Neurosci Ther 2024; 30:e70026. [PMID: 39223758 PMCID: PMC11369019 DOI: 10.1111/cns.70026] [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: 06/19/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
AIMS Incidence of acute mountain sickness (AMS) ranges from 40%-90%, with high-altitude cerebral edema (HACE) representing a life-threatening end stage of severe AMS. However, practical and convenient preventive strategies for HACE are lacking. Remote ischemic preconditioning (RIPC) has demonstrated preventive effects on ischemia- or hypoxia-induced cardiovascular and cerebrovascular diseases. This study aimed to investigate the potential molecular mechanism of HACE and the application of RIPC in preventing HACE onset. METHODS A hypobaric hypoxia chamber was used to simulate a high-altitude environment of 7000 meters. Metabolomics and metabolic flux analysis were employed to assay metabolite levels. Transcriptomics and quantitative real-time PCR (q-PCR) were used to investigate gene expression levels. Immunofluorescence staining was performed on neurons to label cellular proteins. The fluorescent probes Mito-Dendra2, iATPSnFR1.0, and CMTMRos were used to observe mitochondria, ATP, and membrane potential in cultured neurons, respectively. TUNEL staining was performed to detect and quantify apoptotic cell death. Hematoxylin and eosin (H&E) staining was utilized to analyze pathological changes, such as tissue swelling in cerebral cortex samples. The Rotarod test was performed to assess motor coordination and balance in rats. Oxygen-glucose deprivation (OGD) of cultured cells was employed as an in vitro model to simulate the hypoxia and hypoglycemia induced by RIPC in animal experiments. RESULTS We revealed a causative perturbation of glucose metabolism in the brain preceding cerebral edema. Ischemic preconditioning treatment significantly reprograms glucose metabolism, ameliorating cell apoptosis and hypoxia-induced energy deprivation. Notably, ischemic preconditioning improves mitochondrial membrane potential and ATP production through enhanced glucose-coupled mitochondrial metabolism. In vivo studies confirm that RIPC alleviates cerebral edema, reduces cell apoptosis induced by high-altitude hypoxia, and improves motor dysfunction resulting from cerebral edema. CONCLUSIONS Our study elucidates the metabolic basis of HACE pathogenesis. This study provides a new strategy for preventing HACE that RIPC reduces brain edema through reprogramming metabolism, highlighting the potential of targeting metabolic reprogramming for neuroprotective interventions in neurological diseases caused by ischemia or hypoxia.
Collapse
Affiliation(s)
- Rongrong Han
- Beijing Advanced Innovation Center for Big Data‐Based Precision MedicineBeihang UniversityBeijingChina
| | - Xiaoyan Yang
- Beijing Advanced Innovation Center for Big Data‐Based Precision MedicineBeihang UniversityBeijingChina
| | - Xunming Ji
- Beijing Advanced Innovation Center for Big Data‐Based Precision MedicineBeihang UniversityBeijingChina
- China‐America Institute of Neuroscience, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Bing Zhou
- Beijing Advanced Innovation Center for Big Data‐Based Precision MedicineBeihang UniversityBeijingChina
- School of Medical Science and EngineeringBeihang UniversityBeijingChina
| |
Collapse
|
33
|
Albericio G, Higuera M, Araque P, Sánchez C, Herrero D, García-Brenes MA, Formentini L, Torán JL, Mora C, Bernad A. Development of a Bmi1+ Cardiac Mouse Progenitor Immortalized Model to Unravel the Relationship with Its Protective Vascular Endothelial Niche. Int J Mol Sci 2024; 25:8815. [PMID: 39201501 PMCID: PMC11354400 DOI: 10.3390/ijms25168815] [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: 07/03/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
The adult mammalian heart has been demonstrated to be endowed with low but real turnover capacity, especially for cardiomyocytes, the key functional cell type. The source, however, of that turnover capacity remains controversial. In this regard, we have defined and characterized a resident multipotent cardiac mouse progenitor population, Bmi1+DR (for Bmi1+ Damage-Responsive cells). Bmi1+DR is one of the cell types with the lowest ROS (Reactive Oxygen Species) levels in the adult heart, being particularly characterized by their close relationship with cardiac vessels, most probably involved in the regulation of proliferation/maintenance of Bmi1+DR. This was proposed to work as their endothelial niche. Due to the scarcity of Bmi1+DR cells in the adult mouse heart, we have generated an immortalization/dis-immortalization model using Simian Vacuolating Virus 40-Large Antigen T (SV40-T) to facilitate their in vitro characterization. We have obtained a heterogeneous population of immortalized Bmi1+DR cells (Bmi1+DRIMM) that was validated attending to different criteria, also showing a comparable sensitivity to strong oxidative damage. Then, we concluded that the Bmi1-DRIMM population is an appropriate model for primary Bmi1+DR in vitro studies. The co-culture of Bmi1+DRIMM cells with endothelial cells protects them against oxidative damage, showing a moderate depletion in non-canonical autophagy and also contributing with a modest metabolic regulation.
Collapse
Affiliation(s)
- Guillermo Albericio
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
- Molecular Biology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Marina Higuera
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| | - Paula Araque
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| | - Cristina Sánchez
- Molecular Biology Department, Molecular Biology Center Severo Ochoa (CBMSO), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Diego Herrero
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| | - Miguel A. García-Brenes
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| | - Laura Formentini
- Molecular Biology Department, Molecular Biology Center Severo Ochoa (CBMSO), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - José Luis Torán
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| | - Carmen Mora
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| | - Antonio Bernad
- Cardiac Stem Cells Lab, Immunology and Oncology Department, National Center for Biotechnology (CNB-CSIC), Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain; (G.A.); (M.H.); (P.A.); (J.L.T.)
| |
Collapse
|
34
|
Chen Z, Yang C, Ji J, Chen M, Han B. Umbilical Cord Blood-Derived Cells Can Reconstruct Hematopoiesis in an Aplastic Anemia Animal Model. Stem Cells Int 2024; 2024:4095268. [PMID: 39161367 PMCID: PMC11333133 DOI: 10.1155/2024/4095268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/16/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Objectives To explore the efficacy and the mechanism of the umbilical cord-derived cells combined with cyclosporine A (CsA) in treating aplastic anemia (AA) in mice. Methods Immune-mediated AA model mice were treated with CsA + UC mesenchymal stem cells (UC-MSC), CsA + umbilical cord blood regulatory T cells (UCB-Treg), UC-MSC, UCB-Treg, CsA alone, or blank control, respectively (n = 9 mice/group). CsA and the cell infusion was administered on d0. Routine peripheral blood testing was performed once weekly; bone marrow colony culture, bone marrow cell flow cytometry, peripheral blood T cell subsets, and serum inflammatory cytokines tests were performed on d14. Transcriptome sequencing was performed for cells from CsA + UC-MSC, CsA + UCB-Treg, and CsA groups to detect the possible related genes. Gene function cluster and signal pathway enrichment analysis were also performed. Results Blank control mice died due to pancytopenia within 21 days, whereas mice in other groups survived for >28 days. On d14, the CsA + UC-MSC and CsA + UCB-Treg groups had higher white blood cell (WBC) counts than the other groups (p < 0.05), along with higher burst-forming unit (BFU) and colony-forming unit-granulocyte, macrophage (CFU-GM) counts (p < 0.01). The CsA + UC-MSC group had the highest BFU count (p < 0.01). The CsA + UC-MSC and CsA + UCB-Treg groups exhibited the highest bone marrow CD34+ cell proportion (9.68% ± 1.35% and 8.17% ± 0.53%, respectively; p < 0.01). Tumor necrosis factor (TNF)-α and interleukin (IL)-2 levels in the CsA + UC-MSC group (p < 0.05) and TNF-α, interleukin-2, and interferon (INF)-γ levels in the CsA + UC-Treg group (p < 0.01) were lower than those in the CsA group. Compared with CsA treatment, CsA + UC-MSC significantly downregulated the histone methylation pathway (p < 0.05), whereas CsA + UCB-Treg significantly upregulated energy metabolism processes (p < 0.05). Treatment with CsA + UC-MSC upregulated superoxide dismutase activity compared with CsA + UCB-Treg treatment. Conclusions Adding UC-MSC or UCB-Treg to CsA markedly enhanced the reconstruction of hematopoiesis in AA mice, with UC-MSC eliciting greater efficiency than UCB-Treg. Accordingly, the addition of these cells could further improve immune abnormalities.
Collapse
Affiliation(s)
- Zesong Chen
- Department of HematologyPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- Department of OncologyCancer Hospital Chinese Academy of Medical SciencesShenzhen Hospital, Shenzhen, China
| | - Chen Yang
- Department of HematologyPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Jiang Ji
- Department of HematologyPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Miao Chen
- Department of HematologyPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Bing Han
- Department of HematologyPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
35
|
Cázares-Preciado JA, López-Arredondo A, Cruz-Cardenas JA, Luévano-Martínez LA, García-Rivas G, Prado-Garcia H, Brunck MEG. Metabolic features of neutrophilic differentiation of HL-60 cells in hyperglycemic environments. BMJ Open Diabetes Res Care 2024; 12:e004181. [PMID: 39122366 PMCID: PMC11409339 DOI: 10.1136/bmjdrc-2024-004181] [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: 03/08/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
INTRODUCTION Chronic hyperglycemia affects neutrophil functions, leading to reduced pathogen killing and increased morbidity. This impairment has been directly linked to increased glycemia, however, how this specifically affects neutrophils metabolism and their differentiation in the bone marrow is unclear and difficult to study. RESEARCH DESIGN AND METHODS We used high-resolution respirometry to investigate the metabolism of resting and activated donor neutrophils, and flow cytometry to measure surface CD15 and CD11b expression. We then used HL-60 cells differentiated towards neutrophil-like cells in standard media and investigated the effect of doubling glucose concentration on differentiation metabolism. We measured the oxygen consumption rate (OCR), and the enzymatic activity of carnitine palmitoyl transferase 1 (CPT1) and citrate synthase during neutrophil-like differentiation. We compared the surface phenotype, functions, and OCR of neutrophil-like cells differentiated under both glucose concentrations. RESULTS Donor neutrophils showed significant instability of CD11b and OCR after phorbol 12-myristate 13-acetate stimulation at 3 hours post-enrichment. During HL-60 neutrophil-like cell differentiation, there was a significant increase in surface CD15 and CD11b expression together with the loss of mitochondrial mass. Differentiated neutrophil-like cells also exhibited higher CD11b expression and were significantly more phagocytic. In higher glucose media, we measured a decrease in citrate synthase and CPT1 activities during neutrophil-like differentiation. CONCLUSIONS HL-60 neutrophil-like differentiation recapitulated known molecular and metabolic features of human neutrophil differentiation. Increased glucose concentrations correlated with features described in hyperglycemic donor neutrophils including increased CD11b and phagocytosis. We used this model to describe metabolic features of neutrophil-like cell differentiation in hyperglycemia and show for the first time the downregulation of CPT1 and citrate synthase activity, independently of mitochondrial mass.
Collapse
Affiliation(s)
| | | | | | - Luis Alberto Luévano-Martínez
- Escuela de Medicina y Ciencias de La Salud, Tecnologico de Monterrey, Monterrey, Nuevo Leon, Mexico
- The Institute for Obesity Research, Tecnologico de Monterrey, Monterrey, Mexico
| | - Gerardo García-Rivas
- Escuela de Medicina y Ciencias de La Salud, Tecnologico de Monterrey, Monterrey, Nuevo Leon, Mexico
- The Institute for Obesity Research, Tecnologico de Monterrey, Monterrey, Mexico
| | - Heriberto Prado-Garcia
- Laboratorio de Onco-Inmunobiología, Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico, Mexico
| | - Marion E G Brunck
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, Nuevo Leon, Mexico
- The Institute for Obesity Research, Tecnologico de Monterrey, Monterrey, Mexico
| |
Collapse
|
36
|
Park Y, Lee I, Lee MJ, Park H, Jung GS, Kim N, Im W, Kim H, Lee JH, Cho S, Choi YS. Particulate matter exposure induces adverse effects on endometrium and fertility via aberrant inflammatory and apoptotic pathways in vitro and in vivo. CHEMOSPHERE 2024; 361:142466. [PMID: 38810796 DOI: 10.1016/j.chemosphere.2024.142466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/27/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024]
Abstract
This study aimed to evaluate the adverse effects of particulate matter (PM) exposure on endometrial cells and fertility and to identify possible underlying mechanisms. Thirteen women (aged 15-52 years) were included in this study. Enrolled patients underwent laparoscopic surgery at Gangnam Severance Hospital between 1 January and 31 December 2021. For in vivo experiments, 36 female and nine male C57BL/6 mice were randomly divided into control(vehicle), low-dose(10 mg/kg/d), and high-dose exposure groups(20 mg/kg/d). PM was inhaled nasally for four weeks and natural mating was performed. NIST® SRM® 1648a was used for PM exposure. qRT-PCR, western blotting and Masson's trichrome staining were performed. PM treatment in human endometrial stromal cells induced inflammation with significant upregulation of IL-1β, p-NF-kB, and p-c-Jun compared to those of controls. Additionally, PM treatment significantly increased apoptosis in human endometrial stromal cells by downregulating p-AKT and upregulating p-p53/p53, Cas-3, BAX/Bcl-2, p-AMPK, and p-ERK. After PM treatment, the relative expression of IL-1β, IL-6, TNF-α, p-NF-κB, p-c-Jun, and p-Nrf2/Nrf2 significantly increased in murine endometrium compared to those of the controls. Expression of apoptotic proteins p53, p27, and Cas-3, was also significantly elevated in murine endometrium of the PM exposure group compared to that of the controls. A significant increase in expression of procollagen Ⅰ, and Masson's trichrome staining scores in the murine endometrium was noted after PM treatment. PM treatment significantly decreased ERα expression. After natural mating, all 3 female mice in the control group gave birth to 25 offspring (mean 8.1), whereas in the low-dose PM treatment group, two of three female mice gave birth to nine offspring (mean 4.5). No pregnant mice or offspring was present in the high-dose PM treatment group. PM exposure induces adverse effects on the endometrium through aberrant activation of inflammatory and apoptotic pathways and is associated with detrimental effects on murine fertility.
Collapse
Affiliation(s)
- Yunjeong Park
- Department of Obstetrics and Gynecology, Guro Hospital, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Inha Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea; Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Min Jung Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea; Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hyemin Park
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea
| | - Gee Soo Jung
- Department of Medical Device Engineering and Management, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea
| | - Nara Kim
- Department of Medical Device Engineering and Management, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea
| | - Wooseok Im
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea; Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Heeyon Kim
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jae Hoon Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea; Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - SiHyun Cho
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, Republic of Korea; Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Young Sik Choi
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| |
Collapse
|
37
|
Racine L, Parmentier R, Niphadkar S, Chhun J, Martignoles JA, Delhommeau F, Laxman S, Paldi A. Metabolic adaptation pilots the differentiation of human hematopoietic cells. Life Sci Alliance 2024; 7:e202402747. [PMID: 38802246 PMCID: PMC11130395 DOI: 10.26508/lsa.202402747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
A continuous supply of energy is an essential prerequisite for survival and represents the highest priority for the cell. We hypothesize that cell differentiation is a process of optimization of energy flow in a changing environment through phenotypic adaptation. The mechanistic basis of this hypothesis is provided by the established link between core energy metabolism and epigenetic covalent modifications of chromatin. This theory predicts that early metabolic perturbations impact subsequent differentiation. To test this, we induced transient metabolic perturbations in undifferentiated human hematopoietic cells using pharmacological inhibitors targeting key metabolic reactions. We recorded changes in chromatin structure and gene expression, as well as phenotypic alterations by single-cell ATAC and RNA sequencing, time-lapse microscopy, and flow cytometry. Our observations suggest that these metabolic perturbations are shortly followed by alterations in chromatin structure, leading to changes in gene expression. We also show that these transient fluctuations alter the differentiation potential of the cells.
Collapse
Affiliation(s)
- Laëtitia Racine
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- Ecole Pratique des Hautes Etudes, PSL Research University, Paris, France
- AP-HP, SIRIC CURAMUS, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France
- OPALE Carnot Institute, Paris, France
| | - Romuald Parmentier
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- Ecole Pratique des Hautes Etudes, PSL Research University, Paris, France
- AP-HP, SIRIC CURAMUS, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France
- OPALE Carnot Institute, Paris, France
| | - Shreyas Niphadkar
- Institute for Stem Cell Science and Regenerative Medicine (DBT-inStem), Bangalore, India
| | - Julie Chhun
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- Ecole Pratique des Hautes Etudes, PSL Research University, Paris, France
- AP-HP, SIRIC CURAMUS, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France
- OPALE Carnot Institute, Paris, France
| | - Jean-Alain Martignoles
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- AP-HP, SIRIC CURAMUS, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France
- OPALE Carnot Institute, Paris, France
| | - François Delhommeau
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- AP-HP, SIRIC CURAMUS, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France
- OPALE Carnot Institute, Paris, France
| | - Sunil Laxman
- Institute for Stem Cell Science and Regenerative Medicine (DBT-inStem), Bangalore, India
| | - Andras Paldi
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- Ecole Pratique des Hautes Etudes, PSL Research University, Paris, France
- AP-HP, SIRIC CURAMUS, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France
- OPALE Carnot Institute, Paris, France
| |
Collapse
|
38
|
Morganti C, Bonora M, Ito K. Metabolism and HSC fate: what NADPH is made for. Trends Cell Biol 2024:S0962-8924(24)00141-7. [PMID: 39054107 PMCID: PMC11757803 DOI: 10.1016/j.tcb.2024.07.003] [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: 05/01/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
Mitochondrial metabolism plays a central role in the regulation of hematopoietic stem cell (HSC) biology. Mitochondrial fatty acid oxidation (FAO) is pivotal in controlling HSC self-renewal and differentiation. Herein, we discuss recent evidence suggesting that NADPH generated in the mitochondria can influence the fate of HSCs. Although NADPH has multiple functions, HSCs show high levels of NADPH that are preferentially used for cholesterol biosynthesis. Endogenous cholesterol supports the biogenesis of extracellular vesicles (EVs), which are essential for maintaining HSC properties. We also highlight the significance of EVs in hematopoiesis through autocrine signaling. Elucidating the mitochondrial NADPH-cholesterol axis as part of the metabolic requirements of healthy HSCs will facilitate the development of new therapies for hematological disorders.
Collapse
Affiliation(s)
- Claudia Morganti
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA.
| | - Massimo Bonora
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA.
| |
Collapse
|
39
|
Kafeel S, Ragone A, Salzillo A, Palmiero G, Naviglio S, Sapio L. Adiponectin Receptor Agonist AdipoRon Inhibits Proliferation and Drives Glycolytic Dependence in Non-Small-Cell Lung Cancer Cells. Cancers (Basel) 2024; 16:2633. [PMID: 39123363 PMCID: PMC11312309 DOI: 10.3390/cancers16152633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Despite the countless therapeutic advances achieved over the years, non-small-cell lung cancer (NSCLC) is the leading cause of cancer-related death worldwide. To this primacy contribute both non-oncogene addicted and advanced NSCLCs, in which conventional therapies are only partially effective. The adiponectin receptor agonist AdipoRon has revealed antiproliferative action in different cancers, including osteosarcoma and pancreatic cancer. Herein, we investigated its potential anticancer role in NSCLC for the first time. We proved that AdipoRon strongly inhibits viability, growth and colony formation in H1299 and A549 NSCLC cells, mainly through a slowdown in cell cycle progression. Along with the biological behaviors, a metabolic switching was observed after AdipoRon administration in NSCLC cells, consisting of higher glucose consumption and lactate accumulation. Remarkably, both 2-Deoxy Glucose and Oxamate glycolytic-interfering agents greatly enhanced AdipoRon's antiproliferative features. As a master regulator of cell metabolism, AMP-activated protein kinase (AMPK) was activated by AdipoRon. Notably, the ablation of AdipoRon-induced AMPK phosphorylation by Compound-C significantly counteracted its effectiveness. However, the engagement of other pathways should be investigated afterwards. With a focus on NSCLC, our findings further support the ability of AdipoRon in acting as an anticancer molecule, driving its endorsement as a future candidate in NSCLC therapy.
Collapse
Affiliation(s)
| | | | | | | | - Silvio Naviglio
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.K.); (A.R.); (A.S.); (G.P.); (L.S.)
| | | |
Collapse
|
40
|
Wu J, Liu T, Tang M, Liu Y, Wang W, Wang C, Ju Y, Zhao Y, Zhang Y. Ex Vivo Evaluation of the Function of Hematopoietic Stem Cells in Toxicology of Metals. Curr Protoc 2024; 4:e1038. [PMID: 38967962 DOI: 10.1002/cpz1.1038] [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: 07/06/2024]
Abstract
A variety of metals, e.g., lead (Pb), cadmium (Cd), and lithium (Li), are in the environment and are toxic to humans. Hematopoietic stem cells (HSCs) reside at the apex of hematopoiesis and are capable of generating all kinds of blood cells and self-renew to maintain the HSC pool. HSCs are sensitive to environmental stimuli. Metals may influence the function of HSCs by directly acting on HSCs or indirectly by affecting the surrounding microenvironment for HSCs in the bone marrow (BM) or niche, including cellular and extracellular components. Investigating the impact of direct and/or indirect actions of metals on HSCs contributes to the understanding of immunological and hematopoietic toxicology of metals. Treatment of HSCs with metals ex vivo, and the ensuing HSC transplantation assays, are useful for evaluating the impacts of the direct actions of metals on the function of HSCs. Investigating the mechanisms involved, given the rarity of HSCs, methods that require large numbers of cells are not suitable for signal screening; however, flow cytometry is a useful tool for signal screening HSCs. After targeting signaling pathways, interventions ex vivo and HSCs transplantation are required to confirm the roles of the signaling pathways in regulating the function of HSCs exposed to metals. Here, we describe protocols to evaluate the mechanisms of direct and indirect action of metals on HSCs. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Identify the impact of a metal on the competence of HSCs Basic Protocol 2: Identify the impact of a metal on the lineage bias of HSC differentiation Basic Protocol 3: Screen the potential signaling molecules in HSCs during metal exposure Alternate Protocol 1: Ex vivo treatment with a metal on purified HSCs Alternate Protocol 2: Ex vivo intervention of the signaling pathway regulating the function of HSCs during metal exposure.
Collapse
Affiliation(s)
- Jiaojiao Wu
- Experimental Center for Research, School of Public Health, Fudan University, Shanghai, China
| | - Ting Liu
- Experimental Center for Research, School of Public Health, Fudan University, Shanghai, China
| | - Mengke Tang
- Experimental Center for Research, School of Public Health, Fudan University, Shanghai, China
| | - Yalin Liu
- Experimental Center for Research, School of Public Health, Fudan University, Shanghai, China
| | - Wei Wang
- Experimental Center for Research, School of Public Health, Fudan University, Shanghai, China
| | - Chuanxuan Wang
- Experimental Center for Research, School of Public Health, Fudan University, Shanghai, China
| | - Yingzi Ju
- Experimental Center for Research, School of Public Health, Fudan University, Shanghai, China
| | - Yifan Zhao
- Experimental Center for Research, School of Public Health, Fudan University, Shanghai, China
| | - Yubin Zhang
- Experimental Center for Research, School of Public Health, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Jaramillo V, Arévalo DF, González-Hernández M, Cortés MT, Perdomo-Arciniegas AM, Cruz JC, Muñoz-Camargo C. Conductive extracellular matrix derived/chitosan methacrylate/ graphene oxide-pegylated hybrid hydrogel for cell expansion. Front Bioeng Biotechnol 2024; 12:1398052. [PMID: 38952668 PMCID: PMC11215370 DOI: 10.3389/fbioe.2024.1398052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/08/2024] [Indexed: 07/03/2024] Open
Abstract
Electrical stimulation has emerged as a cornerstone technique in the rapidly evolving field of biomedical engineering, particularly within the realms of tissue engineering and regenerative medicine. It facilitates cell growth, proliferation, and differentiation, thereby advancing the development of accurate tissue models and enhancing drug-testing methodologies. Conductive hydrogels, which enable the conduction of microcurrents in 3D in vitro cultures, are central to this advancement. The integration of high-electroconductive nanomaterials, such as graphene oxide (GO), into hydrogels has revolutionized their mechanical and conductivity properties. Here, we introduce a novel electrostimulation assay utilizing a hybrid hydrogel composed of methacryloyl-modified small intestine submucosa (SIS) dECM (SISMA), chitosan methacrylate (ChiMA), and GO-polyethylene glycol (GO-PEG) in a 3D in vitro culture within a hypoxic environment of umbilical cord blood cells (UCBCs). Results not only demonstrate significant cell proliferation within 3D constructs exposed to microcurrents and early growth factors but also highlight the hybrid hydrogel's physiochemical prowess through comprehensive rheological, morphological, and conductivity analyses. Further experiments will focus on identifying the regulatory pathways of cells subjected to electrical stimulation.
Collapse
Affiliation(s)
- Valentina Jaramillo
- Grupo de investigación en Nanobiomateriales, Ingeniería Celular y Bioimpresión (GINIB), Departamento de Ingeniería Biomédica, Universidad de los Andes, Bogotá, Colombia
| | - Daniel Felipe Arévalo
- Grupo de investigación en Nanobiomateriales, Ingeniería Celular y Bioimpresión (GINIB), Departamento de Ingeniería Biomédica, Universidad de los Andes, Bogotá, Colombia
| | | | - María T. Cortés
- Department of Chemistry, Universidad de los Andes, Bogotá, Colombia
| | - Ana María Perdomo-Arciniegas
- Cord Blood Bank (CBB) Research Group, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud (IDCBIS), Bogotá, Colombia
| | - Juan C. Cruz
- Grupo de investigación en Nanobiomateriales, Ingeniería Celular y Bioimpresión (GINIB), Departamento de Ingeniería Biomédica, Universidad de los Andes, Bogotá, Colombia
| | - Carolina Muñoz-Camargo
- Grupo de investigación en Nanobiomateriales, Ingeniería Celular y Bioimpresión (GINIB), Departamento de Ingeniería Biomédica, Universidad de los Andes, Bogotá, Colombia
| |
Collapse
|
42
|
Tian Y, Cheng Z, Ge D, Xu Z, Wang H, Li X, Tian H, Liu F, Luo D, Wang Y. ROS are required for the germinative cell proliferation and metacestode larval growth of Echinococcus multilocularis. Front Microbiol 2024; 15:1410504. [PMID: 38912347 PMCID: PMC11190091 DOI: 10.3389/fmicb.2024.1410504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/24/2024] [Indexed: 06/25/2024] Open
Abstract
The potentially lethal zoonotic disease alveolar echinococcosis (AE) is caused by the metacestode larval stages of the tapeworm Echinococcus multilocularis. Metacestode growth and proliferation occurs within the inner organs of mammalian hosts, which is associated with complex molecular parasite-host interactions. The host has developed various ways to resist a parasitic infection, and the production of reactive oxygen species (ROS) is one of the most important strategies. Here, we found that scavenging of ROS reduced metacestode larval growth and germinative cell proliferation in in vivo models. Furthermore, using in vitro-cultured metacestode vesicles, we found that increased ROS levels enhanced metacestode growth and germinative cell proliferation, which was achieved by positively activating the ROS-EmERK-EmHIF1α axis. These results indicate that, beside its capacity to damage the parasite, ROS also play critical roles in metacestode growth and germinative cell proliferation. This study suggests that the effects of ROS on parasite may be bidirectional during AE infection, reflecting the parasite's adaptation to the oxidative stress microenvironment.
Collapse
Affiliation(s)
- Ye Tian
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhe Cheng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Defeng Ge
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhijian Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Huijuan Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xiazhen Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Huimin Tian
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Fan Liu
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Damin Luo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yanhai Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
43
|
Liao W, Chen X, Zhang S, Chen J, Liu C, Yu K, Zhang Y, Chen M, Chen F, Shen M, Lu B, Han S, Wang S, Wang J, Du C. Megakaryocytic IGF1 coordinates activation and ferroptosis to safeguard hematopoietic stem cell regeneration after radiation injury. Cell Commun Signal 2024; 22:292. [PMID: 38802843 PMCID: PMC11129484 DOI: 10.1186/s12964-024-01651-5] [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] [Received: 02/25/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Hematopoietic stem cell (HSC) regeneration underlies hematopoietic recovery from myelosuppression, which is a life-threatening side effect of cytotoxicity. HSC niche is profoundly disrupted after myelosuppressive injury, while if and how the niche is reshaped and regulates HSC regeneration are poorly understood. METHODS A mouse model of radiation injury-induced myelosuppression was built by exposing mice to a sublethal dose of ionizing radiation. The dynamic changes in the number, distribution and functionality of HSCs and megakaryocytes were determined by flow cytometry, immunofluorescence, colony assay and bone marrow transplantation, in combination with transcriptomic analysis. The communication between HSCs and megakaryocytes was determined using a coculture system and adoptive transfer. The signaling mechanism was investigated both in vivo and in vitro, and was consolidated using megakaryocyte-specific knockout mice and transgenic mice. RESULTS Megakaryocytes become a predominant component of HSC niche and localize closer to HSCs after radiation injury. Meanwhile, transient insulin-like growth factor 1 (IGF1) hypersecretion is predominantly provoked in megakaryocytes after radiation injury, whereas HSCs regenerate paralleling megakaryocytic IGF1 hypersecretion. Mechanistically, HSCs are particularly susceptible to megakaryocytic IGF1 hypersecretion, and mTOR downstream of IGF1 signaling not only promotes activation including proliferation and mitochondrial oxidative metabolism of HSCs, but also inhibits ferritinophagy to restrict HSC ferroptosis. Consequently, the delicate coordination between proliferation, mitochondrial oxidative metabolism and ferroptosis ensures functional HSC expansion after radiation injury. Importantly, punctual IGF1 administration simultaneously promotes HSC regeneration and hematopoietic recovery after radiation injury, representing a superior therapeutic approach for myelosuppression. CONCLUSIONS Our study identifies megakaryocytes as a last line of defense against myelosuppressive injury and megakaryocytic IGF1 as a novel niche signal safeguarding HSC regeneration.
Collapse
Affiliation(s)
- Weinian Liao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Xinliang Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Shuzhen Zhang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Jun Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Chaonan Liu
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Kuan Yu
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Yimin Zhang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Mo Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Fang Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Mingqiang Shen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Binghui Lu
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Songling Han
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Song Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China.
| | - Changhong Du
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
44
|
Branco A, Rayabaram J, Miranda CC, Fernandes-Platzgummer A, Fernandes TG, Sajja S, da Silva CL, Vemuri MC. Advances in ex vivo expansion of hematopoietic stem and progenitor cells for clinical applications. Front Bioeng Biotechnol 2024; 12:1380950. [PMID: 38846805 PMCID: PMC11153805 DOI: 10.3389/fbioe.2024.1380950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/25/2024] [Indexed: 06/09/2024] Open
Abstract
As caretakers of the hematopoietic system, hematopoietic stem cells assure a lifelong supply of differentiated populations that are responsible for critical bodily functions, including oxygen transport, immunological protection and coagulation. Due to the far-reaching influence of the hematopoietic system, hematological disorders typically have a significant impact on the lives of individuals, even becoming fatal. Hematopoietic cell transplantation was the first effective therapeutic avenue to treat such hematological diseases. Since then, key use and manipulation of hematopoietic stem cells for treatments has been aspired to fully take advantage of such an important cell population. Limited knowledge on hematopoietic stem cell behavior has motivated in-depth research into their biology. Efforts were able to uncover their native environment and characteristics during development and adult stages. Several signaling pathways at a cellular level have been mapped, providing insight into their machinery. Important dynamics of hematopoietic stem cell maintenance were begun to be understood with improved comprehension of their metabolism and progressive aging. These advances have provided a solid platform for the development of innovative strategies for the manipulation of hematopoietic stem cells. Specifically, expansion of the hematopoietic stem cell pool has triggered immense interest, gaining momentum. A wide range of approaches have sprouted, leading to a variety of expansion systems, from simpler small molecule-based strategies to complex biomimetic scaffolds. The recent approval of Omisirge, the first expanded hematopoietic stem and progenitor cell product, whose expansion platform is one of the earliest, is predictive of further successes that might arise soon. In order to guarantee the quality of these ex vivo manipulated cells, robust assays that measure cell function or potency need to be developed. Whether targeting hematopoietic engraftment, immunological differentiation potential or malignancy clearance, hematopoietic stem cells and their derivatives need efficient scaling of their therapeutic potency. In this review, we comprehensively view hematopoietic stem cells as therapeutic assets, going from fundamental to translational.
Collapse
Affiliation(s)
- André Branco
- Department of Bioengineering and Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Janakiram Rayabaram
- Protein and Cell Analysis, Biosciences Division, Invitrogen Bioservices, Thermo Fisher Scientific, Bangalore, India
| | - Cláudia C. Miranda
- Department of Bioengineering and Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- AccelBio, Collaborative Laboratory to Foster Translation and Drug Discovery, Cantanhede, Portugal
| | - Ana Fernandes-Platzgummer
- Department of Bioengineering and Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago G. Fernandes
- Department of Bioengineering and Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Suchitra Sajja
- Protein and Cell Analysis, Biosciences Division, Invitrogen Bioservices, Thermo Fisher Scientific, Bangalore, India
| | - Cláudia L. da Silva
- Department of Bioengineering and Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | | |
Collapse
|
45
|
Luanpitpong S, Tangkiettrakul K, Kang X, Srisook P, Poohadsuan J, Samart P, Klaihmon P, Janan M, Lorthongpanich C, Laowtammathron C, Issaragrisil S. OGT and OGA gene-edited human induced pluripotent stem cells for dissecting the functional roles of O-GlcNAcylation in hematopoiesis. Front Cell Dev Biol 2024; 12:1361943. [PMID: 38752196 PMCID: PMC11094211 DOI: 10.3389/fcell.2024.1361943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/14/2024] [Indexed: 05/18/2024] Open
Abstract
Hematopoiesis continues throughout life to produce all types of blood cells from hematopoietic stem cells (HSCs). Metabolic state is a known regulator of HSC self-renewal and differentiation, but whether and how metabolic sensor O-GlcNAcylation, which can be modulated via an inhibition of its cycling enzymes O-GlcNAcase (OGA) and O-GlcNAc transferase (OGT), contributes to hematopoiesis remains largely unknown. Herein, isogenic, single-cell clones of OGA-depleted (OGAi) and OGT-depleted (OGTi) human induced pluripotent stem cells (hiPSCs) were successfully generated from the master hiPSC line MUSIi012-A, which were reprogrammed from CD34+ hematopoietic stem/progenitor cells (HSPCs) containing epigenetic memory. The established OGAi and OGTi hiPSCs exhibiting an increase or decrease in cellular O-GlcNAcylation concomitant with their loss of OGA and OGT, respectively, appeared normal in phenotype and karyotype, and retained pluripotency, although they may favor differentiation toward certain germ lineages. Upon hematopoietic differentiation through mesoderm induction and endothelial-to-hematopoietic transition, we found that OGA inhibition accelerates hiPSC commitment toward HSPCs and that disruption of O-GlcNAc homeostasis affects their commitment toward erythroid lineage. The differentiated HSPCs from all groups were capable of giving rise to all hematopoietic progenitors, thus confirming their functional characteristics. Altogether, the established single-cell clones of OGTi and OGAi hiPSCs represent a valuable platform for further dissecting the roles of O-GlcNAcylation in blood cell development at various stages and lineages of blood cells. The incomplete knockout of OGA and OGT in these hiPSCs makes them susceptible to additional manipulation, i.e., by small molecules, allowing the molecular dynamics studies of O-GlcNAcylation.
Collapse
Affiliation(s)
- Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kantpitchar Tangkiettrakul
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Xing Kang
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pimonwan Srisook
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jirarat Poohadsuan
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Parinya Samart
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Phatchanat Klaihmon
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Montira Janan
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chanchao Lorthongpanich
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chuti Laowtammathron
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
46
|
Asgari R, Mehran YZ, Weber HM, Weber M, Golestanha SA, Hosseini Kazerouni SM, Panahi F, Mohammadi P, Mansouri K. Management of oxidative stress for cell therapy through combinational approaches of stem cells, antioxidants, and photobiomodulation. Eur J Pharm Sci 2024; 196:106715. [PMID: 38301971 DOI: 10.1016/j.ejps.2024.106715] [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: 10/01/2023] [Revised: 01/05/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
Over the recent decades, stem cell-based therapies have been considered as a beneficial approach for the treatment of various diseases. In these types of therapies, the stem cells and their products are used as treating agents. Despite the helpful efficacy of stem cell-based therapies, there may be challenges. Oxidative stress (OS) is one of these challenges that can affect the therapeutic properties of stem cells. Therefore, it seems that employing strategies for the reduction of OS in combination with stem cell therapy can lead to better results of these therapies. Based on the available evidence, antioxidant therapy and photobiomodulation (PBM) are strategies that can regulate the OS in the cells. Antioxidant therapy is a method in which various antioxidants are used in the therapeutic processes. PBM is also the clinical application of light that gained importance in medicine. Antioxidants and PBM can regulate OS by the effect on mitochondria as an important source of OS in the cells. Considering the importance of OS in pathologic pathways and its effect on the treatment outcomes of stem cells, in the present review first the stem cell therapy and effects of OS on this type of therapy are summarized. Then, antioxidant therapy and PBM as approaches for reducing OS with a focus on mitochondrial function are discussed. Also, a novel combination treatment with the hope of achieving better and more stable outcomes in the treatment process of diseases is proposed.
Collapse
Affiliation(s)
- Rezvan Asgari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Yasaman Zandi Mehran
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hans Michael Weber
- International Society of Medical Laser Applications, Lauenfoerde, Germany
| | | | | | | | - Farzad Panahi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
47
|
Xu J, Fei P, Simon DW, Morowitz MJ, Mehta PA, Du W. Crosstalk between DNA Damage Repair and Metabolic Regulation in Hematopoietic Stem Cells. Cells 2024; 13:733. [PMID: 38727270 PMCID: PMC11083014 DOI: 10.3390/cells13090733] [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: 03/23/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
Self-renewal and differentiation are two characteristics of hematopoietic stem cells (HSCs). Under steady physiological conditions, most primitive HSCs remain quiescent in the bone marrow (BM). They respond to different stimuli to refresh the blood system. The transition from quiescence to activation is accompanied by major changes in metabolism, a fundamental cellular process in living organisms that produces or consumes energy. Cellular metabolism is now considered to be a key regulator of HSC maintenance. Interestingly, HSCs possess a distinct metabolic profile with a preference for glycolysis rather than oxidative phosphorylation (OXPHOS) for energy production. Byproducts from the cellular metabolism can also damage DNA. To counteract such insults, mammalian cells have evolved a complex and efficient DNA damage repair (DDR) system to eliminate various DNA lesions and guard genomic stability. Given the enormous regenerative potential coupled with the lifetime persistence of HSCs, tight control of HSC genome stability is essential. The intersection of DDR and the HSC metabolism has recently emerged as an area of intense research interest, unraveling the profound connections between genomic stability and cellular energetics. In this brief review, we delve into the interplay between DDR deficiency and the metabolic reprogramming of HSCs, shedding light on the dynamic relationship that governs the fate and functionality of these remarkable stem cells. Understanding the crosstalk between DDR and the cellular metabolism will open a new avenue of research designed to target these interacting pathways for improving HSC function and treating hematologic disorders.
Collapse
Affiliation(s)
- Jian Xu
- Division of Hematology and Oncology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15232, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Peiwen Fei
- Cancer Biology, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI 96812, USA
| | - Dennis W. Simon
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michael J. Morowitz
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Parinda A. Mehta
- Division of Blood and Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Wei Du
- Division of Hematology and Oncology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15232, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
48
|
Chen N, Xie QM, Song SM, Guo SN, Fang Y, Fei GH, Wu HM. Dexamethasone protects against asthma via regulating Hif-1α-glycolysis-lactate axis and protein lactylation. Int Immunopharmacol 2024; 131:111791. [PMID: 38460304 DOI: 10.1016/j.intimp.2024.111791] [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: 11/14/2023] [Revised: 02/18/2024] [Accepted: 02/29/2024] [Indexed: 03/11/2024]
Abstract
PURPOSE Asthma can not be eradicated till now and its control primarily relies on the application of corticosteroids. Recently, glycolytic reprogramming has been reportedly contributed to asthma, this study aimed to reveal whether the effect of corticosteroids on asthma control is related to their regulation of glycolysis and glycolysis-dependent protein lactylation. METHODS Ovalbumin (OVA) aeroallergen was used to challenge mice and stimulate human macrophage cell line THP-1 following dexamethasone (DEX) treatment. Airway hyperresponsiveness, airway inflammation, the expressions of key glycolytic enzymes and pyroptosis markers, the level of lactic acid, real-time glycolysis and oxidative phosphorylation (OXPHOS), and protein lactylation were analyzed. RESULTS DEX significantly attenuated OVA-induced eosinophilic airway inflammation, including airway hyperresponsiveness, leukocyte infiltration, goblet cell hyperplasia, Th2 cytokines production and pyroptosis markers expression. Meanwhile, OVA-induced Hif-1α-glycolysis axis was substantially downregulated by DEX, which resulted in low level of lactic acid. Besides, key glycolytic enzymes in the lungs of asthmatic mice were notably co-localized with F4/80-positive macrophages, indicating metabolic shift to glycolysis in lung macrophages during asthma. This was confirmed in OVA-stimulated THP-1 cells that DEX treatment resulted in reductions in pyroptosis, glycolysis and lactic acid level. Finally, protein lactylation was found significantly increased in the lungs of asthmatic mice and OVA-stimulated THP-1 cells, which were both inhibited by DEX. CONCLUSION Our present study revealed that the effect of DEX on asthma control was associated with its suppressing of Hif-1α-glycolysis-lactateaxis and subsequent protein lactylation, which may open new avenues for the therapy of eosinophilic asthma.
Collapse
Affiliation(s)
- Ning Chen
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road No. 218, Hefei, Anhui 230022, PR China
| | - Qiu-Meng Xie
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road No. 218, Hefei, Anhui 230022, PR China
| | - Si-Ming Song
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road No. 218, Hefei, Anhui 230022, PR China
| | - Si-Nuo Guo
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road No. 218, Hefei, Anhui 230022, PR China
| | - Yu Fang
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road No. 218, Hefei, Anhui 230022, PR China
| | - Guang-He Fei
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China.
| | - Hui-Mei Wu
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Jixi Road 218, Hefei, Anhui 230022, PR China; Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Jixi Road No. 218, Hefei, Anhui 230022, PR China.
| |
Collapse
|
49
|
Zervopoulou E, Grigoriou M, Doumas SA, Yiannakou D, Pavlidis P, Gasparoni G, Walter J, Filia A, Gakiopoulou H, Banos A, Mitroulis I, Boumpas DT. Enhanced medullary and extramedullary granulopoiesis sustain the inflammatory response in lupus nephritis. Lupus Sci Med 2024; 11:e001110. [PMID: 38471723 DOI: 10.1136/lupus-2023-001110] [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: 11/22/2023] [Accepted: 02/20/2024] [Indexed: 03/14/2024]
Abstract
OBJECTIVES In SLE, deregulation of haematopoiesis is characterised by inflammatory priming and myeloid skewing of haematopoietic stem and progenitor cells (HSPCs). We sought to investigate the role of extramedullary haematopoiesis (EMH) as a key player for tissue injury in systemic autoimmune disorders. METHODS Transcriptomic analysis of bone marrow (BM)-derived HSPCs from patients with SLE and NZBW/F1 lupus-prone mice was performed in combination with DNA methylation profile. Trained immunity (TI) was induced through β-glucan administration to the NZBW/F1 lupus-prone model. Disease activity was assessed through lupus nephritis (LN) histological grading. Colony-forming unit assay and adoptive cell transfer were used to assess HSPCs functionalities. RESULTS Transcriptomic analysis shows that splenic HSPCs carry a higher inflammatory potential compared with their BM counterparts. Further induction of TI, through β-glucan administration, exacerbates splenic EMH, accentuates myeloid skewing and worsens LN. Methylomic analysis of BM-derived HSPCs demonstrates myeloid skewing which is in part driven by epigenetic tinkering. Importantly, transcriptomic analysis of human SLE BM-derived HSPCs demonstrates similar findings to those observed in diseased mice. CONCLUSIONS These data support a key role of granulocytes derived from primed HSPCs both at medullary and extramedullary sites in the pathogenesis of LN. EMH and TI contribute to SLE by sustaining the systemic inflammatory response and increasing the risk for flare.
Collapse
Affiliation(s)
- Eleni Zervopoulou
- Autoimmunity and Inflammation Laboratory, Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
- 4th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Grigoriou
- Autoimmunity and Inflammation Laboratory, Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
- 1st Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace School of Health Sciences, Alexandroupoli, Greece
| | - Stavros A Doumas
- Autoimmunity and Inflammation Laboratory, Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
- 4th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Danae Yiannakou
- Institute of Computer Science, Foundation of Research and Technology Hellas, Heraklion, Greece
| | - Pavlos Pavlidis
- Institute of Computer Science, Foundation of Research and Technology Hellas, Heraklion, Greece
| | - Gilles Gasparoni
- Department of Genetics-Epigenetics, Saarland University, Saarbrucken, Germany
| | - Jörn Walter
- Department of Genetics-Epigenetics, Saarland University, Saarbrucken, Germany
| | - Anastasia Filia
- Autoimmunity and Inflammation Laboratory, Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
- 1st Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace School of Health Sciences, Alexandroupoli, Greece
| | - Harikleia Gakiopoulou
- 1st Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Aggelos Banos
- Autoimmunity and Inflammation Laboratory, Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Ioannis Mitroulis
- 1st Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace School of Health Sciences, Alexandroupoli, Greece
| | - Dimitrios T Boumpas
- Autoimmunity and Inflammation Laboratory, Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
- 4th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
50
|
Potter E, Dolgova E, Proskurina A, Ruzanova V, Efremov Y, Kirikovich S, Oshikhmina S, Mamaev A, Taranov O, Bryukhovetskiy A, Grivtsova L, Kolchanov N, Ostanin A, Chernykh E, Bogachev S. Stimulation of mouse hematopoietic stem cells by angiogenin and DNA preparations. Braz J Med Biol Res 2024; 57:e13072. [PMID: 38451606 PMCID: PMC10913394 DOI: 10.1590/1414-431x2024e13072] [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: 11/23/2023] [Accepted: 01/24/2024] [Indexed: 03/08/2024] Open
Abstract
Immature hematopoietic progenitors are a constant source for renewal of hemocyte populations and the basic component of the tissue and cell repair apparatus. A unique property of these cells of internalizing extracellular double-stranded DNA has been previously shown. The leukostimulatory effect demonstrated in our pioneering studies was considered to be due to the feature of this cell. In the present research, we have analyzed the effects of DNA genome reconstructor preparation (DNAgr), DNAmix, and human recombinant angiogenin on both hematopoietic stem cells and multipotent progenitors. Treatment with bone marrow cells of experimental mice with these preparations stimulates colony formation by hematopoietic stem cells and proliferation of multipotent descendants. The main lineage responsible for this is the granulocyte-macrophage hematopoietic lineage. Using fluorescent microscopy as well as FACS assay, co-localization of primitive c-Kit- and Sca-1-positive progenitors and the TAMRA-labeled double-stranded DNA has been shown. Human recombinant angiogenin was used as a reference agent. Cells with specific markers were quantified in intact bone marrow and colonies grown in the presence of inducers. Quantitative analysis revealed that a total of 14,000 fragment copies of 500 bp, which is 0.2% of the haploid genome, can be delivered into early progenitors. Extracellular double-stranded DNA fragments stimulated the colony formation in early hematopoietic progenitors from the bone marrow, which assumed their effect on cells in G0. The observed number of Sca1+/c-Kit+ cells in colonies testifies to the possibility of both symmetrical and asymmetrical division of the initial hematopoietic stem cell and its progeny.
Collapse
Affiliation(s)
- E.A. Potter
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E.V. Dolgova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A.S. Proskurina
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - V.S. Ruzanova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Y.R. Efremov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk National Research State University, Novosibirsk, Russia
| | - S.S. Kirikovich
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - S.G. Oshikhmina
- Department of Natural Sciences, Novosibirsk National Research State University, Novosibirsk, Russia
| | - A.L. Mamaev
- LLC “Angiopharm Laboratory”, Novosibirsk, Russia
| | - O.S. Taranov
- State Research Center of Virology and Biotechnology “Vector”, Novosibirsk, Russia
| | | | - L.U. Grivtsova
- Department of Clinical Immunology, National Medical Research Radiological Centre, Ministry of Health of the Russian Federation, Obninsk, Russia
| | - N.A. Kolchanov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A.A. Ostanin
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - E.R. Chernykh
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - S.S. Bogachev
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|