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
|
Hadjimichael E, Deitsch KW. Variable surface antigen expression, virulence, and persistent infection by Plasmodium falciparum malaria parasites. Microbiol Mol Biol Rev 2025; 89:e0011423. [PMID: 39807932 PMCID: PMC11948492 DOI: 10.1128/mmbr.00114-23] [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: 01/16/2025] Open
Abstract
SUMMARYThe human malaria parasite Plasmodium falciparum is known for its ability to maintain lengthy infections that can extend for over a year. This property is derived from the parasite's capacity to continuously alter the antigens expressed on the surface of the infected red blood cell, thereby avoiding antibody recognition and immune destruction. The primary target of the immune system is an antigen called PfEMP1 that serves as a cell surface receptor and enables infected cells to adhere to the vascular endothelium and thus avoid filtration by the spleen. The parasite's genome encodes approximately 60 antigenically distinct forms of PfEMP1, each encoded by individual members of the multicopy var gene family. This provides the parasite with a repertoire of antigenic types that it systematically cycles through over the course of an infection, thereby maintaining an infection until the repertoire is exhausted. While this model of antigenic variation based on var gene switching explains the dynamics of acute infections in individuals with limited anti-malarial immunity, it fails to explain reports of chronic, asymptomatic infections that can last over a decade. Recent field studies have led to a re-evaluation of previous conclusions regarding the prevalence of chronic infections, and the application of new technologies has provided insights into the molecular mechanisms that enable chronic infections and how these processes evolved.
Collapse
Affiliation(s)
- Evi Hadjimichael
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
| | - Kirk W. Deitsch
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
3
|
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
|
4
|
Daumova L, Manakov D, Petrak J, Sovilj D, Behounek M, Andera L, Vit O, Souckova O, Havranek O, Dolnikova A, Renesova N, Tuskova L, Winkowska L, Bettazova N, Kupcova K, Kalbacova MH, Sikorova M, Trneny M, Klener P. Long-term adaptation of lymphoma cell lines to hypoxia is mediated by diverse molecular mechanisms that are targetable with specific inhibitors. Cell Death Discov 2025; 11:65. [PMID: 39966387 PMCID: PMC11836139 DOI: 10.1038/s41420-025-02341-y] [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: 08/05/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
A large body of evidence suggests that hypoxia drives aggressive molecular features of malignant cells irrespective of cancer type. Non-Hodgkin lymphomas (NHL) are the most common hematologic malignancies characterized by frequent involvement of diverse hypoxic microenvironments. We studied the impact of long-term deep hypoxia (1% O2) on the biology of lymphoma cells. Only 2 out of 6 tested cell lines (Ramos, and HBL2) survived ≥ 4 weeks under hypoxia. The hypoxia-adapted (HA)b Ramos and HBL2 cells had a decreased proliferation rate accompanied by significant suppression of both oxidative phosphorylation and glycolytic pathways. Transcriptome and proteome analyses revealed marked downregulation of genes and proteins of the mitochondrial respiration complexes I and IV, and mitochondrial ribosomal proteins. Despite the observed suppression of glycolysis, the proteome analysis of both HA cell lines showed upregulation of several proteins involved in the regulation of glucose utilization including the active catalytic component of prolyl-4-hydroxylase P4HA1, an important druggable oncogene. HA cell lines demonstrated increased transcription of key regulators of auto-/mitophagy, e.g., neuritin, BCL2 interacting protein 3 (BNIP3), BNIP3-like protein, and BNIP3 pseudogene. Adaptation to hypoxia was further associated with deregulation of apoptosis, namely upregulation of BCL2L1/BCL-XL, overexpression of BCL2L11/BIM, increased binding of BIM to BCL-XL, and significantly increased sensitivity of both HA cell lines to A1155463, a BCL-XL inhibitor. Finally, in both HA cell lines AKT kinase was hyperphosphorylated and the cells showed increased sensitivity to copanlisib, a pan-PI3K inhibitor. In conclusion, our data report on several shared mechanisms of lymphoma cell adaptation to long-term hypoxia including: 1. Upregulation of proteins responsible for glucose utilization, 2. Degradation of mitochondrial proteins for potential mitochondrial recycling (by mitophagy), and 3. Increased dependence on BCL-XL and PI3K-AKT signaling for survival. In translation, inhibition of glycolysis, BCL-XL, or PI3K-AKT cascade may result in targeted elimination of HA lymphoma cells.
Collapse
Affiliation(s)
- Lenka Daumova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Dmitry Manakov
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jiri Petrak
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Dana Sovilj
- Institute of Biotechnology, Czech Academy of Sciences / BIOCEV, Vestec, Czech Republic
| | - Matej Behounek
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Ladislav Andera
- Institute of Biotechnology, Czech Academy of Sciences / BIOCEV, Vestec, Czech Republic
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Ondrej Vit
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Olga Souckova
- OMICS Mass Spectrometry Core Facility, Biology Departments, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Ondrej Havranek
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alex Dolnikova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Nicol Renesova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Liliana Tuskova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lucie Winkowska
- CLIP, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Nardjas Bettazova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kristyna Kupcova
- BIOCEV Biotechnology and Biomedicine Centre, First Faculty of Medicine, Charles University, Vestec, Czech Republic
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marie Hubalek Kalbacova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Miriama Sikorova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marek Trneny
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pavel Klener
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
- First Department of Medicine- Hematology, University General Hospital Prague and First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
5
|
Yuan F, Liu J, Zhong L, Liu P, Li T, Yang K, Gao W, Zhang G, Sun J, Zou X. Enhanced therapeutic effects of hypoxia-preconditioned mesenchymal stromal cell-derived extracellular vesicles in renal ischemic injury. Stem Cell Res Ther 2025; 16:39. [PMID: 39901252 PMCID: PMC11792194 DOI: 10.1186/s13287-025-04166-z] [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: 10/16/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) secreted by mesenchymal stromal cells (MSCs) have been shown to provide significant protection against renal ischemia-reperfusion injury (IRI). Hypoxia has emerged as a promising strategy to enhance the tissue repair capabilities of MSCs. However, the specific effects of hypoxia on MSCs and MSC-EVs, as well as their therapeutic potential in renal IRI, remain unclear. In this study, we investigated the alterations occurring in MSCs and the production of MSC-EVs following hypoxia pre-treatment, and further explored the key intrinsic mechanisms underlying the therapeutic effects of hypoxic MSC-EVs in the treatment of renal IRI. METHODS Human umbilical cord MSCs were cultured under normoxic and hypoxic conditions. Proliferation and related pathways were measured, and RNA sequencing was used to detect changes in the transcriptional profile. MSC-EVs from both normoxic and hypoxic conditions were isolated and characterized. In vivo, the localization and therapeutic effects of MSC-EVs were assessed in a rat renal IRI model. Histological examinations were conducted to evaluate the structure, proliferation, and apoptosis of IRI kidney tissue respectively. Renal function was assessed by measuring serum creatinine and blood urea nitrogen levels. In vitro, the therapeutic potential of MSC-EVs were measured in renal tubular epithelial cells injured by antimycin A. Protein sequencing analysis of hypoxic MSC-EVs was performed, and the depletion of Glutathione S-Transferase Omega 1 (GSTO1) in hypoxic MSC-EVs was carried out to verify its key role in alleviating renal injury. RESULTS Hypoxia alters MSCs transcriptional profile, promotes their proliferation, and increases the production of EVs. Hypoxia-pretreated MSC-EVs demonstrated a superior ability to mitigate renal IRI, enhancing proliferation and reducing apoptosis of renal tubular epithelial cells both in vivo and in vitro. Protein profiling of the EVs revealed an accumulation of numerous anti-oxidative stress proteins, with GSTO1 being particularly prominent. Knockdown of GSTO1 significantly reduced the antioxidant and therapeutic effects on renal IRI of hypoxic MSC-EVs. CONCLUSIONS Hypoxia significantly promotes the generation of MSC-EVs and enhances their therapeutic effects on renal IRI. The antioxidant stress effect induced by GSTO1 is identified as one of the most critical underlying mechanisms. Our findings highlight that hypoxia-pretreated MSC-EVs represent a novel and promising therapeutic strategy for renal IRI.
Collapse
Affiliation(s)
- Fei Yuan
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jie Liu
- Department of Neurology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Zhong
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Pengtao Liu
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ting Li
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Kexin Yang
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Wei Gao
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Guangyuan Zhang
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu Province, China.
| | - Jie Sun
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Xiangyu Zou
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
6
|
Aldridge DL, Lanzar Z, Phan AT, Christian DA, Pardy R, Min B, Kedl RM, Hunter CA. IL-27 limits HSPC differentiation during infection and protects from stem cell exhaustion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633135. [PMID: 39868131 PMCID: PMC11761129 DOI: 10.1101/2025.01.15.633135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Many inflammatory stimuli can induce progenitor cells in the bone marrow to produce increased numbers of myeloid cells as part of the process of emergency myelopoiesis. These events are associated with innate training and can have long-term impacts on hematopoietic stem and progenitor cell (HSPC) development but can also compromise their function. While many cytokines support emergency myelopoiesis, less is known about the mechanisms that temper these events. When mice that lack the cytokine IL-27 were infected with Toxoplasma gondii, there was enhanced generation of monocyte progenitors and increased numbers of inflammatory monocytes. In the bone marrow of infected mice there was increased production of IL-27 that localized with HSPCs and a survey of cytokine receptor expression highlighted that HSPCs were uniquely poised to respond to IL-27. Furthermore, the use of in vitro differentiation assays and mixed bone marrow chimeras revealed that HSPCs from IL-27 deficient mice are pre-disposed towards the monocyte lineage. Additional studies highlighted that after infection loss of the IL-27R resulted in reduced HSPC fitness that manifested as reduced proliferative responses and a decreased ability to reconstitute the hematopoietic system. Thus, the ability of IL-27 to act on HSPC provides a regulatory brake on differentiation to limit monocyte induction and preserve HSPC stemness.
Collapse
Affiliation(s)
- Daniel L Aldridge
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Zachary Lanzar
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Anthony T Phan
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - David A Christian
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Ryan Pardy
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Booki Min
- Department of Microbiology and Immunology, Feinber School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ross M Kedl
- University of Colorado, Anschuitz Medical Campus, Aurora, CO, USA
| | | |
Collapse
|
7
|
Liu S, Vivona ES, Kurre P. Why hematopoietic stem cells fail in Fanconi anemia: Mechanisms and models. Bioessays 2025; 47:e2400191. [PMID: 39460396 DOI: 10.1002/bies.202400191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/27/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
Fanconi anemia (FA) is generally classified as a DNA repair disorder, conferring a genetic predisposition to cancer and prominent bone marrow failure (BMF) in early childhood. Corroborative human and murine studies point to a fetal origin of hematopoietic stem cell (HSC) attrition under replicative stress. Along with intriguing recent insights into non-canonical roles and domain-specific functions of FA proteins, these studies have raised the possibility of a DNA repair-independent BMF etiology. However, deeper mechanistic insight is critical as current curative options of allogeneic stem cell transplantation and emerging gene therapy have limited eligibility, carry significant side effects, and involve complex procedures restricted to resource-rich environments. To develop rational and broadly accessible therapies for FA patients, the field will need more faithful disease models that overcome the scarcity of patient samples, leverage technological advances, and adopt investigational clinical trial designs tailored for rare diseases.
Collapse
Affiliation(s)
- Suying Liu
- Comprehensive Bone Marrow Failure Center, Children's Hospital of Philadelphia, Philadelphia, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - E S Vivona
- Comprehensive Bone Marrow Failure Center, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Peter Kurre
- Comprehensive Bone Marrow Failure Center, Children's Hospital of Philadelphia, Philadelphia, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
8
|
Pruitt L, Abbott RK. Hypoxia-adenosinergic regulation of B cell responses. Front Immunol 2024; 15:1478506. [PMID: 39559353 PMCID: PMC11570280 DOI: 10.3389/fimmu.2024.1478506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/15/2024] [Indexed: 11/20/2024] Open
Abstract
Hypoxic microenvironments induce widespread metabolic changes that have been shown to be critical in regulating innate and adaptive immune responses. Hypoxia-induced changes include the generation of extracellular adenosine followed by subsequent signaling through adenosine receptors on immune cells. This evolutionarily conserved "hypoxia-adenosinergic" pathway of hypoxia → extracellular adenosine → adenosine receptor signaling has been shown to be critical in limiting and redirecting T cell responses including in tumor microenvironments and the gut mucosa. However, the question of whether hypoxic microenvironments are involved in the development of B cell responses has remained unexplored until recently. The discovery that germinal centers (GC), the anatomic site in which B cells undergo secondary diversification and affinity maturation, develop a hypoxic microenvironment has sparked new interest in how this evolutionarily conserved pathway affects antibody responses. In this review we will summarize what is known about hypoxia-adenosinergic microenvironments in lymphocyte development and ongoing immune responses. Specific focus will be placed on new developments regarding the role of the hypoxia-adenosinergic pathway in regulating GC development and humoral immunity.
Collapse
Affiliation(s)
| | - Robert K. Abbott
- Department of Pathology, University of Texas Medical Branch,
Galveston, TX, United States
| |
Collapse
|
9
|
Salwa A, Ferraresi A, Vallino L, Maheshwari C, Moia R, Gaidano G, Isidoro C. High Mitophagy and Low Glycolysis Predict Better Clinical Outcomes in Acute Myeloid Leukemias. Int J Mol Sci 2024; 25:11527. [PMID: 39519080 PMCID: PMC11546612 DOI: 10.3390/ijms252111527] [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: 09/20/2024] [Revised: 10/20/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Acute myeloid leukemia (AML) emerges as one of the most common and fatal leukemias. Treatment of the disease remains highly challenging owing to profound metabolic rewiring mechanisms that confer plasticity to AML cells, ultimately resulting in therapy resistance. Autophagy, a highly conserved lysosomal-driven catabolic process devoted to macromolecular turnover, displays a dichotomous role in AML by suppressing or promoting disease development and progression. Glycolytic metabolism represents a pivotal strategy for AML cells to sustain increasing energy needs related to uncontrolled growth during disease progression. In this study, we tested the hypothesis that a high glycolytic rate and low autophagy flux could represent an advantage for AML cell proliferation and thus be detrimental for patient's prognosis, and vice versa. TCGA in silico analysis of the AML cohort shows that the high expression of MAP1LC3B (along with that of BECN1 and with low expression of p62/SQSTM1) and the high expression of BNIP3 (along with that of PRKN and of MAP1LC3B), which together are indicative of increased autophagy and mitophagy, correlate with better prognosis. On the other hand, the high expression of glycolytic markers HK2, PFKM, and PKM correlates with poor prognosis. Most importantly, the association of a low expression of glycolytic markers with a high expression of autophagy-mitophagy markers conferred the longest overall survival for AML patients. Transcriptomic analysis showed that this combined signature correlates with the downregulation of a subset of genes required for the differentiation of myeloid cells, lactate/pyruvate transporters, and cell cycle progression, in parallel with the upregulation of genes involved in autophagy/lysosomal trafficking and proteolysis, anti-tumor responses like beta-interferon production, and positive regulation of programmed cell death. Taken together, our data support the view that enhanced autophagy-mitophagy flux together with low glycolytic rate predisposes AML patients to a better clinical outcome, suggesting that autophagy inducers and glucose restrictors may hold potential as adjuvant therapeutics for improving AML management.
Collapse
Affiliation(s)
- Amreen Salwa
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (A.S.); (A.F.); (L.V.); (C.M.)
| | - Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (A.S.); (A.F.); (L.V.); (C.M.)
| | - Letizia Vallino
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (A.S.); (A.F.); (L.V.); (C.M.)
| | - Chinmay Maheshwari
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (A.S.); (A.F.); (L.V.); (C.M.)
| | - Riccardo Moia
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy;
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy;
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via P. Solaroli 17, 28100 Novara, Italy; (A.S.); (A.F.); (L.V.); (C.M.)
| |
Collapse
|
10
|
Burzi IS, Parchi PD, Barachini S, Pardini E, Sardo Infirri G, Montali M, Petrini I. Hypoxia Promotes the Stemness of Mesangiogenic Progenitor Cells and Prevents Osteogenic but not Angiogenic Differentiation. Stem Cell Rev Rep 2024; 20:1830-1842. [PMID: 38914791 PMCID: PMC11457687 DOI: 10.1007/s12015-024-10749-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 06/26/2024]
Abstract
The stem cell niche in the bone marrow is a hypoxic environment, where the low oxygen tension preserves the pluripotency of stem cells. We have identified mesangiogenic progenitor cells (MPC) exhibiting angiogenic and mesenchymal differentiation capabilities in vitro. The effect of hypoxia on MPC has not been previously explored. In this study, MPCs were isolated from volunteers' bone marrow and cultured under both normoxic and hypoxic conditions (3% O2). MPCs maintained their characteristic morphology and surface marker expression (CD18 + CD31 + CD90-CD73-) under hypoxia. However, hypoxic conditions led to reduced MPC proliferation in primary cultures and hindered their differentiation into mesenchymal stem cells (MSCs) upon exposure to differentiative medium. First passage MSCs derived from MPC appeared unaffected by hypoxia, exhibiting no discernible differences in proliferative potential or cell cycle. However, hypoxia impeded the subsequent osteogenic differentiation of MSCs, as evidenced by decreased hydroxyapatite deposition. Conversely, hypoxia did not impact the angiogenic differentiation potential of MPCs, as demonstrated by spheroid-based assays revealing comparable angiogenic sprouting and tube-like formation capabilities under both hypoxic and normoxic conditions. These findings indicate that hypoxia preserves the stemness phenotype of MPCs, inhibits their differentiation into MSCs, and hampers their osteogenic maturation while leaving their angiogenic potential unaffected. Our study sheds light on the intricate effects of hypoxia on bone marrow-derived MPCs and their differentiation pathways.
Collapse
Affiliation(s)
- Irene Sofia Burzi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy
| | - Paolo Domenico Parchi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy
| | - Serena Barachini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56125, Pisa, Italy
| | - Eleonora Pardini
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy
| | - Gisella Sardo Infirri
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy
| | - Marina Montali
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56125, Pisa, Italy
| | - Iacopo Petrini
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Via Savi 2, 56125, Pisa, Italy.
| |
Collapse
|
11
|
Grammer C, Komorowska JA, Swann JB. Vhl safeguards thymic epithelial cell identity and thymopoietic capacity by constraining Hif1a activity during development. iScience 2024; 27:110258. [PMID: 39040069 PMCID: PMC11261450 DOI: 10.1016/j.isci.2024.110258] [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: 07/12/2022] [Revised: 03/15/2024] [Accepted: 06/10/2024] [Indexed: 07/24/2024] Open
Abstract
The thymus is a physiologically hypoxic organ and fulfills its role of generating T cells under low-oxygen conditions. We have therefore investigated how thymic epithelial cells (TECs) cope with physiological hypoxia by focusing on the role of the Hif1a-Vhl axis. In most cell types, the oxygen-labile transcriptional regulator Hif1a is a central player in co-ordinating responses to low oxygen: under normoxic conditions Hif1a is rapidly degraded in a Vhl-guided manner; however, under hypoxic conditions Hif1a is stabilized and can execute its transcriptional functions. Unexpectedly, we find that, although TECs reside in a hypoxic microenvironment, they express little Hif1a protein and do not require Hif1a for their development or function. Instead, we find that Vhl function in TECs is vital to constrain Hif1a activity, as loss of Vhl results in dramatic defects in TEC differentiation and thymopoiesis, which can be rescued by Hif1a co-depletion.
Collapse
Affiliation(s)
- Christiane Grammer
- Department of Developmental Immunology, Max Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Julia A. Komorowska
- Department of Developmental Immunology, Max Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Albert Ludwig University, Faculty of Biology, Freiburg, Germany
| | - Jeremy B. Swann
- Department of Developmental Immunology, Max Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
| |
Collapse
|
12
|
Kapadia CD, Goodell MA. Tissue mosaicism following stem cell aging: blood as an exemplar. NATURE AGING 2024; 4:295-308. [PMID: 38438628 DOI: 10.1038/s43587-024-00589-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/07/2024] [Indexed: 03/06/2024]
Abstract
Loss of stem cell regenerative potential underlies aging of all tissues. Somatic mosaicism, the emergence of cellular patchworks within tissues, increases with age and has been observed in every organ yet examined. In the hematopoietic system, as in most tissues, stem cell aging through a variety of mechanisms occurs in lockstep with the emergence of somatic mosaicism. Here, we draw on insights from aging hematopoiesis to illustrate fundamental principles of stem cell aging and somatic mosaicism. We describe the generalizable changes intrinsic to aged stem cells and their milieu that provide the backdrop for somatic mosaicism to emerge. We discuss genetic and nongenetic mechanisms that can result in tissue somatic mosaicism and existing methodologies to detect such clonal outgrowths. Finally, we propose potential avenues to modify mosaicism during aging, with the ultimate aim of increasing tissue resiliency.
Collapse
Affiliation(s)
- Chiraag D Kapadia
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Margaret A Goodell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
13
|
Jackson BT, Finley LWS. Metabolic regulation of the hallmarks of stem cell biology. Cell Stem Cell 2024; 31:161-180. [PMID: 38306993 PMCID: PMC10842269 DOI: 10.1016/j.stem.2024.01.003] [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: 12/03/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 02/04/2024]
Abstract
Stem cells perform many different functions, each of which requires specific metabolic adaptations. Over the past decades, studies of pluripotent and tissue stem cells have uncovered a range of metabolic preferences and strategies that correlate with or exert control over specific cell states. This review aims to describe the common themes that emerge from the study of stem cell metabolism: (1) metabolic pathways supporting stem cell proliferation, (2) metabolic pathways maintaining stem cell quiescence, (3) metabolic control of cellular stress responses and cell death, (4) metabolic regulation of stem cell identity, and (5) metabolic requirements of the stem cell niche.
Collapse
Affiliation(s)
- Benjamin T Jackson
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, New York, NY, USA
| | - Lydia W S Finley
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
14
|
Zhang C, Ye W, Zhao M, Xia D, Fan Z. tRNA-derived small RNA changes in bone marrow stem cells under hypoxia and osteogenic conduction. J Oral Rehabil 2023; 50:1487-1497. [PMID: 37574812 DOI: 10.1111/joor.13566] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/04/2023] [Accepted: 08/01/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND Tissue engineering using bone mesenchymal stem cells (BMSCs) transplantation is a promising therapeutic for bone regeneration. However, the effect of bone regeneration remains unsatisfactory due to the BMSCs' functional abnormality influenced by hypoxia. In this study, we attempt to explore the mechanism of osteogenic differentiation of BMSCs under hypoxic conditions from the perspective of non-coding RNA regulation. METHODS The study employed BMSCs obtained from healthy donors and simulated hypoxia using CoCl2 stimulation. High-throughput sequencing technique was used to identify differential expression profiles of tRNA-derived small RNA (tsRNA) in three experimental groups: BMSCs-0d, BMSCs-7d and BMSCs-0d-CoCl2 . TargetScan and miRanda algorithms were used to determine tsRNA target genes, while Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis were employed for the prediction of biological functions. Real-time reverse transcriptase-polymerase chain reaction (Real-time RT-PCR) was carried out on four selected differentially expressed tsRNAs. RESULTS After the osteogenic induction and CoCl2 stimulated separately, there were 19 tsRNAs differentially expressed in BMSCs, including 14 upregulated and five downregulated. According to the analysis of biological information, these tsRNAs may regulate 311 potential target genes and mainly enrich the pathways such as metabolic pathways, Wnt signalling pathway, osteoclast differentiation, cellular senescence and mTOR signalling pathway. The results of Real-time RT-PCR for 3'tiRNA-41-GlnTTG-6, 3'tiRNA-42-LysTTT-8, 5'tiRNA-35-CysACA-1 and tRF3a-AsnGTT-9 were consistent with small RNA sequencing data. CONCLUSION We discovered the tsRNA that changes the process of osteogenesis and hypoxia, which provides new targets for promoting survival and regeneration functions after BMSCs transplantation.
Collapse
Affiliation(s)
- Chen Zhang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
- Department of Dental Emergency, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Weilong Ye
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Mengyao Zhao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Dengsheng Xia
- Department of Dental Emergency, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China
- Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Nakahara R, Aki S, Sugaya M, Hirose H, Kato M, Maeda K, Sakamoto DM, Kojima Y, Nishida M, Ando R, Muramatsu M, Pan M, Tsuchida R, Matsumura Y, Yanai H, Takano H, Yao R, Sando S, Shibuya M, Sakai J, Kodama T, Kidoya H, Shimamura T, Osawa T. Hypoxia activates SREBP2 through Golgi disassembly in bone marrow-derived monocytes for enhanced tumor growth. EMBO J 2023; 42:e114032. [PMID: 37781951 PMCID: PMC10646561 DOI: 10.15252/embj.2023114032] [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: 03/15/2023] [Revised: 08/22/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023] Open
Abstract
Bone marrow-derived cells (BMDCs) infiltrate hypoxic tumors at a pre-angiogenic state and differentiate into mature macrophages, thereby inducing pro-tumorigenic immunity. A critical factor regulating this differentiation is activation of SREBP2-a well-known transcription factor participating in tumorigenesis progression-through unknown cellular mechanisms. Here, we show that hypoxia-induced Golgi disassembly and Golgi-ER fusion in monocytic myeloid cells result in nuclear translocation and activation of SREBP2 in a SCAP-independent manner. Notably, hypoxia-induced SREBP2 activation was only observed in an immature lineage of bone marrow-derived cells. Single-cell RNA-seq analysis revealed that SREBP2-mediated cholesterol biosynthesis was upregulated in HSCs and monocytes but not in macrophages in the hypoxic bone marrow niche. Moreover, inhibition of cholesterol biosynthesis impaired tumor growth through suppression of pro-tumorigenic immunity and angiogenesis. Thus, our findings indicate that Golgi-ER fusion regulates SREBP2-mediated metabolic alteration in lineage-specific BMDCs under hypoxia for tumor progression.
Collapse
Affiliation(s)
- Ryuichi Nakahara
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
- Department of Chemistry and Biotechnology, Graduate School of EngineeringThe University of TokyoTokyoJapan
| | - Sho Aki
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
- Department of Chemistry and Biotechnology, Graduate School of EngineeringThe University of TokyoTokyoJapan
| | - Maki Sugaya
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Haruka Hirose
- Department of Systems Biology, Graduate School of MedicineNagoya UniversityNagoyaJapan
- Present address:
Department of Computational and Systems Biology, Medical Research InstituteTokyo Medical and Dental UniversityTokyoJapan
| | - Miki Kato
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Keisuke Maeda
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Daichi M Sakamoto
- Department of Chemistry and Biotechnology, Graduate School of EngineeringThe University of TokyoTokyoJapan
| | - Yasuhiro Kojima
- Department of Systems Biology, Graduate School of MedicineNagoya UniversityNagoyaJapan
| | - Miyuki Nishida
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Ritsuko Ando
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Masashi Muramatsu
- Division of Molecular and Vascular Biology, IRDAKumamoto UniversityKumamotoJapan
| | - Melvin Pan
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Rika Tsuchida
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | | | - Hideyuki Yanai
- Department of Inflammology, RCASTThe University of TokyoTokyoJapan
| | - Hiroshi Takano
- Department of Cell BiologyJapanese Foundation for Cancer ResearchTokyoJapan
| | - Ryoji Yao
- Department of Cell BiologyJapanese Foundation for Cancer ResearchTokyoJapan
| | - Shinsuke Sando
- Department of Chemistry and Biotechnology, Graduate School of EngineeringThe University of TokyoTokyoJapan
- Department of Bioengineering, Graduate School of EngineeringThe University of TokyoTokyoJapan
| | - Masabumi Shibuya
- Institute of Physiology and MedicineJobu UniversityTakasakiJapan
| | - Juro Sakai
- Division of Metabolic Medicine, RCASTThe University of TokyoTokyoJapan
- Division of Molecular Physiology and Metabolism, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Tatsuhiko Kodama
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
| | - Hiroyasu Kidoya
- Department of Signal Transduction, RIMDOsaka UniversityOsakaJapan
- Department of Integrative Vascular Biology, Faculty of Medical SciencesUniversity of FukuiFukuiJapan
| | - Teppei Shimamura
- Department of Systems Biology, Graduate School of MedicineNagoya UniversityNagoyaJapan
- Present address:
Department of Computational and Systems Biology, Medical Research InstituteTokyo Medical and Dental UniversityTokyoJapan
| | - Tsuyoshi Osawa
- Division of Nutriomics and Oncology, RCASTThe University of TokyoTokyoJapan
- Department of Chemistry and Biotechnology, Graduate School of EngineeringThe University of TokyoTokyoJapan
| |
Collapse
|
16
|
Ramachandra N, Gupta M, Schwartz L, Todorova T, Shastri A, Will B, Steidl U, Verma A. Role of IL8 in myeloid malignancies. Leuk Lymphoma 2023; 64:1742-1751. [PMID: 37467070 DOI: 10.1080/10428194.2023.2232492] [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: 03/10/2023] [Revised: 06/06/2023] [Accepted: 06/25/2023] [Indexed: 07/21/2023]
Abstract
Aberrant overexpression of Interleukin-8 (IL8) has been reported in Myelodysplastic Syndromes (MDS), Acute Myeloid Leukemia (AML), Myeloproliferative Neoplasms (MPNs) and other myeloid malignancies. IL8 (CXCL8) is a CXC chemokine that is secreted by aberrant hematopoietic stem and progenitors as well as other cells in the tumor microenvironment. IL8 can bind to CXCR1/CXCR2 receptors and activate oncogenic signaling pathways, and also increase the recruitment of myeloid derived suppressor cells to the tumor microenvironment. IL8/CXCR1/2 overexpression has been associated with poorer prognosis in MDS and AML and increased bone marrow fibrosis in Myelofibrosis. Preclinical studies have demonstrated benefit of inhibiting the IL8/CXCR1/2 pathways via restricting the growth of leukemic stem cells as well as normalizing the immunosuppressive microenvironment in tumors. Targeting the IL8-CXCR1/2 pathway is a potential therapeutic strategy in myeloid neoplasms and is being evaluated with small molecule inhibitors as well as monoclonal antibodies in ongoing clinical trials. We review the role of IL8 signaling pathway in myeloid cancers and discuss future directions on therapeutic targeting of IL8 in these diseases.
Collapse
Affiliation(s)
- Nandini Ramachandra
- Department of Oncology, Blood Cancer Institute, Montefiore Einstein Cancer Center, Bronx, NY, USA
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Malini Gupta
- Department of Cell Biology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Leya Schwartz
- Department of Oncology, Blood Cancer Institute, Montefiore Einstein Cancer Center, Bronx, NY, USA
| | - Tihomira Todorova
- Department of Cell Biology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Aditi Shastri
- Department of Oncology, Blood Cancer Institute, Montefiore Einstein Cancer Center, Bronx, NY, USA
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Britta Will
- Department of Oncology, Blood Cancer Institute, Montefiore Einstein Cancer Center, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Ulrich Steidl
- Department of Oncology, Blood Cancer Institute, Montefiore Einstein Cancer Center, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Amit Verma
- Department of Oncology, Blood Cancer Institute, Montefiore Einstein Cancer Center, Bronx, NY, USA
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|
17
|
Xu Y, Chiang YH, Ho PC, Vannini N. Mitochondria Dictate Function and Fate of HSCs and T Cells. Cancer Immunol Res 2023; 11:1303-1313. [PMID: 37789763 DOI: 10.1158/2326-6066.cir-22-0685] [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: 08/27/2022] [Revised: 01/23/2023] [Accepted: 08/16/2023] [Indexed: 10/05/2023]
Abstract
Hematopoietic stem cells (HSC) and T cells are intimately related, lineage-dependent cell populations that are extensively used as therapeutic products for the treatment of hematologic malignancies and certain types of solid tumors. These cellular therapies can be life-saving treatments; however, their efficacies are often limited by factors influencing their activity and cellular properties. Among these factors is mitochondrial metabolism, which influences the function and fate commitment of both HSCs and T cells. Mitochondria, besides being the "cellular powerhouse," provide metabolic intermediates that are used as substrates for epigenetic modifications and chromatin remodeling, thus, driving cell fate decisions during differentiation. Moreover, mitochondrial fitness and mitochondrial quality control mechanisms are closely related to cellular function, and impairment of these mitochondrial properties associates with cellular dysfunction due to factors such as T-cell exhaustion and aging. Here, we give an overview of the role of mitochondria in shaping the behavior of these lineage-related cell populations. Moreover, we discuss the potential of novel mitochondria-targeting strategies for enhancing HSC- and T cell-based cancer immunotherapies and highlight how design and application of such approaches requires consideration of the metabolic similarities and differences between HSCs and T cells. See related article on p. 1302.
Collapse
Affiliation(s)
- Yingxi Xu
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Yi-Hsuan Chiang
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Nicola Vannini
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
18
|
Şoroğlu CV, Uslu-Bıçak İ, Toprak SF, Yavuz AS, Sözer S. Effect of hypoxia on HIF-1α and NOS3 expressions in CD34 + cells of JAK2V617F-positive myeloproliferative neoplasms. Adv Med Sci 2023; 68:169-175. [PMID: 37075583 DOI: 10.1016/j.advms.2023.03.003] [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: 09/15/2022] [Revised: 02/20/2023] [Accepted: 03/25/2023] [Indexed: 04/21/2023]
Abstract
PURPOSE Myeloproliferative neoplasms (MPN) are a heterogeneous group of hematopoietic stem-cell diseases with excessive proliferation of one or more blood cell lines. In this study, we evaluated the effect of different oxygen concentrations on HIF-1α and NOS3 gene expression to determine the effect of the bone marrow microenvironment on JAK2V617F positive Philadelphia chromosome negative (Ph-) MPNs. PATIENTS AND METHODS Peripheral blood mononuclear cells (MNC) of 12 patients with Ph- MPN were collected. The presence of JAK2V617F allele status was determined with allele-specific nested PCR analysis. MPN CD34+ and CD34depleted populations were isolated from MNC by magnetic beads. Separate cell cultures of CD34+/depleted populations were managed at different oxygen concentrations including anoxia (∼0%), hypoxia (∼3%), and normoxia (∼20%) conditions for 24 h. HIF-1α and NOS3 gene expression changes were examined in each population related to JAK2V617F status with real time RT-PCR. RESULT It was revealed that relative HIF-1α and NOS3 expressions were significantly increased in response to decreased oxygen concentration in all samples. Relative HIF-1α and NOS3 expressions were found to be higher especially in CD34+ and CD34depleted populations carrying JAK2V617F mutations compared to MPN patients carrying wild-type JAK2. CONCLUSION JAK2V617F might have specific role in HIF-1α and NOS3 regulations with respect to low oxygen concentrations in Ph- MPN. Further evaluations might reveal the effect of JAK2V617F on Ph- MPN pathogenesis in bone marrow microenvironment.
Collapse
Affiliation(s)
- Can Veysel Şoroğlu
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Institute of Health Sciences, Istanbul University, Istanbul, Turkey
| | - İldeniz Uslu-Bıçak
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Institute of Health Sciences, Istanbul University, Istanbul, Turkey
| | - Selin Fulya Toprak
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Institute of Health Sciences, Istanbul University, Istanbul, Turkey
| | - Akif Selim Yavuz
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Selçuk Sözer
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
19
|
Ribatti D, d'Amati A. Hematopoiesis and Mast Cell Development. Int J Mol Sci 2023; 24:10679. [PMID: 37445862 DOI: 10.3390/ijms241310679] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Hematopoietic stem cells (HSCs) are defined based on their capacity to replenish themselves (self-renewal) and give rise to all mature hematopoietic cell types (multi-lineage differentiation) over their lifetime. HSCs are mainly distributed in the bone marrow during adult life, harboring HSC populations and a hierarchy of different kinds of cells contributing to the "niche" that supports HSC regulation, myelopoiesis, and lymphopoiesis. In addition, HSC-like progenitors, innate immune cell precursors such as macrophages, mast cells, natural killer cells, innate lymphoid cells, and megakaryocytes and erythrocyte progenitor cells are connected by a series of complex ontogenic relationships. The first source of mast cells is the extraembryonic yolk sac, on embryonic day 7. Mast cell progenitors circulate and enter peripheral tissues where they complete their differentiation. Embryonic mast cell populations are gradually replaced by definitive stem cell-derived progenitor cells. Thereafter, mast cells originate from the bone marrow, developing from the hematopoietic stem cells via multipotent progenitors, common myeloid progenitors, and granulocyte/monocyte progenitors. In this review article, we summarize the knowledge on mast cell sources, particularly focusing on the complex and multifaceted mechanisms intervening between the hematopoietic process and the development of mast cells.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Translational Biomedicine and Neuroscience, School of Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Antonio d'Amati
- Department of Translational Biomedicine and Neuroscience, School of Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| |
Collapse
|
20
|
Wu T, Pelus LM, Plett PA, Sampson CH, Chua HL, Fisher A, Feng H, Liu L, Li H, Ortiz M, Chittajallu S, Luo Q, Bhatwadekar AD, Meyer TB, Zhang X, Zhou D, Fischer KD, McKinzie DL, Miller SJ, Orschell CM. Further Characterization of Multi-Organ DEARE and Protection by 16,16 Dimethyl Prostaglandin E2 in a Mouse Model of the Hematopoietic Acute Radiation Syndrome. Radiat Res 2023; 199:468-489. [PMID: 37014943 PMCID: PMC10278147 DOI: 10.1667/rade-22-00208.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/15/2023] [Indexed: 04/06/2023]
Abstract
Survivors of acute radiation exposure suffer from the delayed effects of acute radiation exposure (DEARE), a chronic condition affecting multiple organs, including lung, kidney, heart, gastrointestinal tract, eyes, and brain, and often causing cancer. While effective medical countermeasures (MCM) for the hematopoietic-acute radiation syndrome (H-ARS) have been identified and approved by the FDA, development of MCM for DEARE has not yet been successful. We previously documented residual bone marrow damage (RBMD) and progressive renal and cardiovascular DEARE in murine survivors of H-ARS, and significant survival efficacy of 16,16-dimethyl prostaglandin E2 (dmPGE2) given as a radioprotectant or radiomitigator for H-ARS. We now describe additional DEARE (physiological and neural function, progressive fur graying, ocular inflammation, and malignancy) developing after sub-threshold doses in our H-ARS model, and detailed analysis of the effects of dmPGE2 administered before (PGE-pre) or after (PGE-post) lethal total-body irradiation (TBI) on these DEARE. Administration of PGE-pre normalized the twofold reduction of white blood cells (WBC) and lymphocytes seen in vehicle-treated survivors (Veh), and increased the number of bone marrow (BM) cells, splenocytes, thymocytes, and phenotypically defined hematopoietic progenitor cells (HPC) and hematopoietic stem cells (HSC) to levels equivalent to those in non-irradiated age-matched controls. PGE-pre significantly protected HPC colony formation ex vivo by >twofold, long term-HSC in vivo engraftment potential up to ninefold, and significantly blunted TBI-induced myeloid skewing. Secondary transplantation documented continued production of LT-HSC with normal lineage differentiation. PGE-pre reduced development of DEARE cardiovascular pathologies and renal damage; prevented coronary artery rarefication, blunted progressive loss of coronary artery endothelia, reduced inflammation and coronary early senescence, and blunted radiation-induced increase in blood urea nitrogen (BUN). Ocular monocytes were significantly lower in PGE-pre mice, as was TBI-induced fur graying. Increased body weight and decreased frailty in male mice, and reduced incidence of thymic lymphoma were documented in PGE-pre mice. In assays measuring behavioral and cognitive functions, PGE-pre reduced anxiety in females, significantly blunted shock flinch response, and increased exploratory behavior in males. No effect of TBI was observed on memory in any group. PGE-post, despite significantly increasing 30-day survival in H-ARS and WBC and hematopoietic recovery, was not effective in reducing TBI-induced RBMD or any other DEARE. In summary, dmPGE2 administered as an H-ARS MCM before lethal TBI significantly increased 30-day survival and ameliorated RBMD and multi-organ and cognitive/behavioral DEARE to at least 12 months after TBI, whereas given after TBI, dmPGE2 enhances survival from H-ARS but has little impact on RBMD or other DEARE.
Collapse
Affiliation(s)
- Tong Wu
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Louis M. Pelus
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - P. Artur Plett
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Carol H. Sampson
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Hui Lin Chua
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Alexa Fisher
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Hailin Feng
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Liqiong Liu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Hongge Li
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Miguel Ortiz
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Supriya Chittajallu
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Qianyi Luo
- Department of Ophthalmology, and Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Ashay D. Bhatwadekar
- Department of Ophthalmology, and Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Timothy B. Meyer
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Xin Zhang
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida 32611
| | - Daohong Zhou
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida 32611
| | - Kathryn D. Fischer
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - David L. McKinzie
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Steven J. Miller
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Christie M. Orschell
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| |
Collapse
|
21
|
Dausinas Ni P, Hartman M, Slack J, Basile C, Liu S, Wan J, O'Leary HA. Novel differential calcium regulation of hematopoietic stem and progenitor cells under physiological low oxygen conditions. J Cell Physiol 2023. [PMID: 37051890 DOI: 10.1002/jcp.30942] [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: 03/14/2022] [Revised: 11/28/2022] [Accepted: 12/23/2022] [Indexed: 04/14/2023]
Abstract
Low oxygen bone marrow (BM) niches (~1%-4% low O2 ) provide critical signals for hematopoietic stem/progenitor cells (HSC/HSPCs). Our presented data are the first to investigate live, sorted HSC/HSPCs in their native low O2 conditions. Transcriptional and proteomic analysis uncovered differential Ca2+ regulation that correlated with overlapping phenotypic populations consisting of robust increases of cytosolic and mitochondrial Ca2+ , ABC transporter (ABCG2) expression and sodium/hydrogen exchanger (NHE1) expression in live, HSC/HSPCs remaining in constant low O2. We identified a novel Ca2+ high population in HSPCs predominantly detected in low O2 that displayed enhanced frequency of phenotypic LSK/LSKCD150 in low O2 replating assays compared to Ca2+ low populations. Inhibition of the Ca2+ regulator NHE1 (Cariporide) resulted in attenuation of both the low O2 induced Ca2+ high population and subsequent enhanced maintenance of phenotypic LSK and LSKCD150 during low O2 replating. These data reveal multiple levels of differential Ca2+ regulation in low O2 resulting in phenotypic, signaling, and functional consequences in HSC/HSPCs.
Collapse
Affiliation(s)
- Paige Dausinas Ni
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Melissa Hartman
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jacob Slack
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Christopher Basile
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Center of Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Center of Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Heather A O'Leary
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| |
Collapse
|
22
|
Tang X, Wang Z, Wang J, Cui S, Xu R, Wang Y. Functions and regulatory mechanisms of resting hematopoietic stem cells: a promising targeted therapeutic strategy. Stem Cell Res Ther 2023; 14:73. [PMID: 37038215 PMCID: PMC10088186 DOI: 10.1186/s13287-023-03316-5] [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: 10/10/2022] [Accepted: 03/29/2023] [Indexed: 04/12/2023] Open
Abstract
Hematopoietic stem cells (HSCs) are the common and essential precursors of all blood cells, including immune cells, and they are responsible for the lifelong maintenance and damage repair of blood tissue homeostasis. The vast majority (> 95%) of HSCs are in a resting state under physiological conditions and are only activated to play a functional role under stress conditions. This resting state affects their long-term survival and is also closely related to the lifelong maintenance of hematopoietic function; however, abnormal changes may also be an important factor leading to the decline of immune function in the body and the occurrence of diseases in various systems. While the importance of resting HSCs has attracted increasing research attention, our current understanding of this topic remains insufficient, and the direction of clinical targeted treatments is unclear. Here, we describe the functions of HSCs, analyze the regulatory mechanisms that affect their resting state, and discuss the relationship between resting HSCs and different diseases, with a view to providing guidance for the future clinical implementation of related targeted treatments.
Collapse
Affiliation(s)
- Xinyu Tang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhenzhen Wang
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingyi Wang
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Siyuan Cui
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruirong Xu
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, China.
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, China.
- Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yan Wang
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, China.
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, China.
- Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
23
|
Mian SA, Philippe C, Maniati E, Protopapa P, Bergot T, Piganeau M, Nemkov T, Bella DD, Morales V, Finch AJ, D’Alessandro A, Bianchi K, Wang J, Gallipoli P, Kordasti S, Kubasch AS, Cross M, Platzbecker U, Wiseman DH, Bonnet D, Bernard DG, Gribben JG, Rouault-Pierre K. Vitamin B5 and succinyl-CoA improve ineffective erythropoiesis in SF3B1-mutated myelodysplasia. Sci Transl Med 2023; 15:eabn5135. [PMID: 36857430 PMCID: PMC7614516 DOI: 10.1126/scitranslmed.abn5135] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/08/2023] [Indexed: 03/03/2023]
Abstract
Patients with myelodysplastic syndrome and ring sideroblasts (MDS-RS) present with symptomatic anemia due to ineffective erythropoiesis that impedes their quality of life and increases morbidity. More than 80% of patients with MDS-RS harbor splicing factor 3B subunit 1 (SF3B1) mutations, the founder aberration driving MDS-RS disease. Here, we report how mis-splicing of coenzyme A synthase (COASY), induced by mutations in SF3B1, affects heme biosynthesis and erythropoiesis. Our data revealed that COASY was up-regulated during normal erythroid differentiation, and its silencing prevented the formation of erythroid colonies, impeded erythroid differentiation, and precluded heme accumulation. In patients with MDS-RS, loss of protein due to COASY mis-splicing led to depletion of both CoA and succinyl-CoA. Supplementation with COASY substrate (vitamin B5) rescued CoA and succinyl-CoA concentrations in SF3B1mut cells and mended erythropoiesis differentiation defects in MDS-RS primary patient cells. Our findings reveal a key role of the COASY pathway in erythroid maturation and identify upstream and downstream metabolites of COASY as a potential treatment for anemia in patients with MDS-RS.
Collapse
Affiliation(s)
- Syed A Mian
- The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Céline Philippe
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Eleni Maniati
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Pantelitsa Protopapa
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Tiffany Bergot
- University of Brest, Inserm, EFS, UMR1078, GGB, 29238 Brest, France
| | | | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Doriana Di Bella
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Valle Morales
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Andrew J Finch
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Katiuscia Bianchi
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Jun Wang
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Paolo Gallipoli
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Shahram Kordasti
- System Cancer Immunology, Comprehensive Cancer Centre, King's College London, London WC2R 2LS, United Kingdom
| | - Anne Sophie Kubasch
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, 04103 Leipzig, Germany
| | - Michael Cross
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, 04103 Leipzig, Germany
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, 04103 Leipzig, Germany
| | - Daniel H Wiseman
- Division of Cancer Sciences, The University of Manchester, Manchester M20 4GJ, UK
| | | | - Delphine G Bernard
- University of Brest, Inserm, EFS, UMR1078, GGB, 29238 Brest, France
- Centre de Ressources Biologiques du CHRU de Brest, 29238 Brest, France
| | - John G Gribben
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Kevin Rouault-Pierre
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| |
Collapse
|
24
|
Zhang J, Han Y, Song M, Wang Q, Cao Z, Yang X, Li Y. Selenium Improves Bone Microenvironment-Related Hematopoiesis and Immunity in T-2 Toxin-Exposed Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:2590-2599. [PMID: 36693005 DOI: 10.1021/acs.jafc.2c08275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The T-2 toxin is one of the most frequent contaminants in the environment and agricultural production globally. It exerts a wide range of toxic effects. Selenium (Se), as an antioxidant, has the potential to be widely used to antagonize mycotoxin toxicity. To investigate the protective effects of Se on bone microenvironment (BM)-related hematopoiesis and immunity after T-2 toxin exposure, 36 male mice were treated with the T-2 toxin (1 mg/kg) and/or Se (0.2 mg/kg) by intragastric administration for 28 days. The results showed that Se alleviated T-2 toxin-induced cytopenia and splenic extramedullary hematopoiesis. Se also significantly relieved T-2 toxin-induced immunosuppression, as assessed by immune factors and lymphocytes. Furthermore, Se also attenuated oxidative stress and apoptosis and improved the BM in T-2 toxin-exposed mice. Therefore, Se improves BM-related hematopoiesis and immunity after T-2 toxin exposure. This study provides references for identifying the toxic mechanism and screening potential therapeutic drugs of the T-2 toxin.
Collapse
Affiliation(s)
- Jian Zhang
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Yanfei Han
- Liaoning Agricultural Technical College, Yingkou, Liaoning 115009, China
| | - Miao Song
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Qi Wang
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Zheng Cao
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Xu Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450002, China
| | - Yanfei Li
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| |
Collapse
|
25
|
Stelmach P, Trumpp A. Leukemic stem cells and therapy resistance in acute myeloid leukemia. Haematologica 2023; 108:353-366. [PMID: 36722405 PMCID: PMC9890038 DOI: 10.3324/haematol.2022.280800] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Indexed: 02/02/2023] Open
Abstract
A major obstacle in the treatment of acute myeloid leukemia (AML) is refractory disease or relapse after achieving remission. The latter arises from a few therapy-resistant cells within minimal residual disease (MRD). Resistant cells with long-term self-renewal capacity that drive clonal outgrowth are referred to as leukemic stem cells (LSC). The cancer stem cell concept considers LSC as relapse-initiating cells residing at the top of each genetically defined AML subclone forming epigenetically controlled downstream hierarchies. LSC display significant phenotypic and epigenetic plasticity, particularly in response to therapy stress, which results in various mechanisms mediating treatment resistance. Given the inherent chemotherapy resistance of LSC, targeted strategies must be incorporated into first-line regimens to prevent LSC-mediated AML relapse. The combination of venetoclax and azacitidine is a promising current strategy for the treatment of AML LSC. Nevertheless, the selection of patients who would benefit either from standard chemotherapy or venetoclax + azacitidine treatment in first-line therapy has yet to be established and the mechanisms of resistance still need to be discovered and overcome. Clinical trials are currently underway that investigate LSC susceptibility to first-line therapies. The era of single-cell multi-omics has begun to uncover the complex clonal and cellular architectures and associated biological networks. This should lead to a better understanding of the highly heterogeneous AML at the inter- and intra-patient level and identify resistance mechanisms by longitudinal analysis of patients' samples. This review discusses LSC biology and associated resistance mechanisms, potential therapeutic LSC vulnerabilities and current clinical trial activities.
Collapse
Affiliation(s)
- Patrick Stelmach
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance,Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM, gGmbH),Department of Medicine V, Heidelberg University Hospital
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM, gGmbH); Faculty of Biosciences, Heidelberg University; German Cancer Consortium (DKTK), Heidelberg.
| |
Collapse
|
26
|
Zeng X, Wang YP, Man CH. Metabolism in Hematopoiesis and Its Malignancy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1442:45-64. [PMID: 38228958 DOI: 10.1007/978-981-99-7471-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Hematopoietic stem cells (HSCs) are multipotent stem cells that can self-renew and generate all blood cells of different lineages. The system is under tight control in order to maintain a precise equilibrium of the HSC pool and the effective production of mature blood cells to support various biological activities. Cell metabolism can regulate different molecular activities, such as epigenetic modification and cell cycle regulation, and subsequently affects the function and maintenance of HSC. Upon malignant transformation, oncogenic drivers in malignant hematopoietic cells can remodel the metabolic pathways for supporting the oncogenic growth. The dysregulation of metabolism results in oncogene addiction, implying the development of malignancy-specific metabolism-targeted therapy. In this chapter, we will discuss the significance of different metabolic pathways in hematopoiesis, specifically, the distinctive metabolic dependency in hematopoietic malignancies and potential metabolic therapy.
Collapse
Affiliation(s)
- Xiaoyuan Zeng
- Division of Haematology, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yi-Ping Wang
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Cheuk-Him Man
- Division of Haematology, Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
27
|
Urao N, Liu J, Takahashi K, Ganesh G. Hematopoietic Stem Cells in Wound Healing Response. Adv Wound Care (New Rochelle) 2022; 11:598-621. [PMID: 34353116 PMCID: PMC9419985 DOI: 10.1089/wound.2021.0065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Significance: Emerging evidence has shown a link between the status of hematopoietic stem cells (HSCs) and wound healing responses. Thus, better understanding HSCs will contribute to further advances in wound healing research. Recent Advances: Myeloid cells such as neutrophils and monocyte-derived macrophages are critical players in the process of wound healing. HSCs actively respond to wound injury and other tissue insults, including infection and produce the effector myeloid cells, and a failing of the HSC response can result in impaired wound healing. Technological advances such as transcriptome at single-cell resolution, epigenetics, three-dimensional imaging, transgenic animals, and animal models, have provided novel concepts of myeloid generation (myelopoiesis) from HSCs, and have revealed cell-intrinsic and -extrinsic mechanisms that can impact HSC functions in the context of health conditions. Critical Issues: The newer concepts include-the programmed cellular fate at a differentiation stage that is used to be considered as the multilineage, the signaling pathways that can activate HSCs directly and indirectly, the mechanisms that can deteriorate HSCs, the roles and remodeling of the surrounding environment for HSCs and their progenitors (the niche). Future Directions: The researches on HSCs, which produce blood cells, should contribute to the development of blood biomarkers predicting a risk of chronic wounds, which may transform clinical practice of wound care with precision medicine for patients at high risk of poor healing.
Collapse
Affiliation(s)
- Norifumi Urao
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, New York, USA.,Correspondence: Department of Pharmacology, State University of New York Upstate Medical University, 766 Irving Avenue, Weiskotten Hall Room 5322, Syracuse, NY 13210, USA.
| | - Jinghua Liu
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Kentaro Takahashi
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Gayathri Ganesh
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
28
|
Arif T. Lysosomes and Their Role in Regulating the Metabolism of Hematopoietic Stem Cells. BIOLOGY 2022; 11:1410. [PMID: 36290314 PMCID: PMC9598322 DOI: 10.3390/biology11101410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/26/2022]
Abstract
Hematopoietic stem cells (HSCs) have the capacity to renew blood cells at all stages of life and are largely quiescent at a steady state. It is essential to understand the processes that govern quiescence in HSCs to enhance bone marrow transplantation. It is hypothesized that in their quiescent state, HSCs primarily use glycolysis for energy production rather than mitochondrial oxidative phosphorylation (OXPHOS). In addition, the HSC switch from quiescence to activation occurs along a continuous developmental path that is driven by metabolism. Specifying the metabolic regulation pathway of HSC quiescence will provide insights into HSC homeostasis for therapeutic application. Therefore, understanding the metabolic demands of HSCs at a steady state is key to developing innovative hematological therapeutics. Lysosomes are the major degradative organelle in eukaryotic cells. Catabolic, anabolic, and lysosomal function abnormalities are connected to an expanding list of diseases. In recent years, lysosomes have emerged as control centers of cellular metabolism, particularly in HSC quiescence, and essential regulators of cell signaling have been found on the lysosomal membrane. In addition to autophagic processes, lysosomal activities have been shown to be crucial in sustaining quiescence by restricting HSCs access to a nutritional reserve essential for their activation into the cell cycle. Lysosomal activity may preserve HSC quiescence by altering glycolysis-mitochondrial biogenesis. The understanding of HSC metabolism has significantly expanded over the decade, revealing previously unknown requirements of HSCs in both their dividing (active) and quiescent states. Therefore, understanding the role of lysosomes in HSCs will allow for the development of innovative treatment methods based on HSCs to fight clonal hematopoiesis and HSC aging.
Collapse
Affiliation(s)
- Tasleem Arif
- Department of Cell, Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
29
|
Elhossaini H, Hamad M, Irhimeh MR, Nakhla S, Rajarathnam GP, Abbas A. Combined hypoxia hypercapnia delays apoptosis and maintains CD34 cell surface antigen. Biochem Eng J 2022. [DOI: 10.1016/j.bej.2022.108650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
30
|
Magliulo D, Bernardi R. Hypoxic stress and hypoxia-inducible factors in leukemias. Front Oncol 2022; 12:973978. [PMID: 36059690 PMCID: PMC9435438 DOI: 10.3389/fonc.2022.973978] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022] Open
Abstract
To cope with hypoxic stress, ancient organisms have developed evolutionally conserved programs centered on hypoxia-inducible transcriptional factors (HIFs). HIFs and their regulatory proteins have evolved as rheostats to adapt cellular metabolism to atmospheric oxygen fluctuations, but the amplitude of their transcriptional programs has tremendously increased along evolution to include a wide spectrum of physiological and pathological processes. The bone marrow represents a notable example of an organ that is physiologically exposed to low oxygen levels and where basal activation of hypoxia signaling appears to be intrinsically wired within normal and neoplastic hematopoietic cells. HIF-mediated responses are mainly piloted by the oxygen-labile α subunits HIF1α and HIF2α, and current literature suggests that these genes have a functional specification that remains to be fully defined. Since their identification in the mid 90s, HIF factors have been extensively studied in solid tumors, while their implication in leukemia has lagged behind. In the last decades however, many laboratories have addressed the function of hypoxia signaling in leukemia and obtained somewhat contradictory results. Suppression of HIFs expression in different types of leukemia has unveiled common leukemia-promoting functions such as stimulation of bone marrow neoangiogenesis, maintenance of leukemia stem cells and chemoresistance. However, genetic studies are revealing that a definition of HIF factors as bona fide tumor promoters is overly simplistic, and, depending on the leukemia subtype, the specific oncogenic event, or the stage of leukemia development, activation of hypoxia-inducible genes may lead to opposite consequences. With this article we will provide an updated summary of the studies describing the regulation and function of HIF1α and HIF2α in blood malignancies, spanning from acute to chronic, lymphoid to myeloid leukemias. In discussing these data, we will attempt to provide plausible explanations to contradictory findings and point at what we believe are areas of weakness in which further investigations are urgently needed. Gaining additional knowledge into the role of hypoxia signaling in leukemia appears especially timely nowadays, as new inhibitors of HIF factors are entering the clinical arena for specific types of solid tumors but their utility for patients with leukemia is yet to be determined.
Collapse
Affiliation(s)
| | - Rosa Bernardi
- Laboratory of Preclinical Models of Cancer, Division of Experimental Oncology, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
31
|
Provenzano A, Chetta M, De Filpo G, Cantini G, La Barbera A, Nesi G, Santi R, Martinelli S, Rapizzi E, Luconi M, Maggi M, Mannelli M, Ercolino T, Canu L. Novel Germline PHD2 Variant in a Metastatic Pheochromocytoma and Chronic Myeloid Leukemia, but in the Absence of Polycythemia. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58081113. [PMID: 36013579 PMCID: PMC9416477 DOI: 10.3390/medicina58081113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/11/2022] [Accepted: 08/14/2022] [Indexed: 11/16/2022]
Abstract
Background: Pheochromocytoma (Pheo) and paraganglioma (PGL) are rare tumors, mostly resulting from pathogenic variants of predisposing genes, with a genetic contribution that now stands at around 70%. Germline variants account for approximately 40%, while the remaining 30% is attributable to somatic variants. Objective: This study aimed to describe a new PHD2 (EGLN1) variant in a patient affected by metastatic Pheo and chronic myeloid leukemia (CML) without polycythemia and to emphasize the need to adopt a comprehensive next-generation sequencing (NGS) panel. Methods: Genetic analysis was carried out by NGS. This analysis was initially performed using a panel of genes known for tumor predisposition (EGLN1, EPAS1, FH, KIF1Bβ, MAX, NF1, RET, SDHA, SDHAF2, SDHB, SDHC, SDHD, TMEM127, and VHL), followed initially by SNP-CGH array, to exclude the presence of the pathogenic Copy Number Variants (CNVs) and the loss of heterozygosity (LOH) and subsequently by whole exome sequencing (WES) comparative sequence analysis of the DNA extracted from tumor fragments and peripheral blood. Results: We found a novel germline PHD2 (EGLN1) gene variant, c.153G>A, p.W51*, in a patient affected by metastatic Pheo and chronic myeloid leukemia (CML) in the absence of polycythemia. Conclusions: According to the latest guidelines, it is mandatory to perform genetic analysis in all Pheo/PGL cases regardless of phenotype. In patients with metastatic disease and no evidence of polycythemia, we propose testing for PHD2 (EGLN1) gene variants. A possible correlation between PHD2 (EGLN1) pathogenic variants and CML clinical course should be considered.
Collapse
Affiliation(s)
- Aldesia Provenzano
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Massimiliano Chetta
- Medical Genetics, Azienda Ospedaliera di Rilievo Nazionale (A.O.R.N.) Cardarelli, Padiglione, 80131 Naples, Italy
| | - Giuseppina De Filpo
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Giulia Cantini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
| | - Andrea La Barbera
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Gabriella Nesi
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Raffaella Santi
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Serena Martinelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
| | - Elena Rapizzi
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy
| | - Michaela Luconi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
| | - Mario Maggi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
- Endocrinology Unit, Azienda Ospedaliera-Universitaria Careggi, 50139 Florence, Italy
| | - Massimo Mannelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
| | - Tonino Ercolino
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
- Endocrinology Unit, Azienda Ospedaliera-Universitaria Careggi, 50139 Florence, Italy
| | - Letizia Canu
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
- Centro di Ricerca e Innovazione sulle Patologie Surrenaliche, AOU Careggi, 50139 Florence, Italy
- European Network for the Study of Adrenal Tumors (ENS@T) Center of Excellence, 50139 Florence, Italy
- Endocrinology Unit, Azienda Ospedaliera-Universitaria Careggi, 50139 Florence, Italy
- Correspondence:
| |
Collapse
|
32
|
Zhang J, Wu X, Ma J, Long K, Sun J, Li M, Ge L. Hypoxia and hypoxia-inducible factor signals regulate the development, metabolism, and function of B cells. Front Immunol 2022; 13:967576. [PMID: 36045669 PMCID: PMC9421003 DOI: 10.3389/fimmu.2022.967576] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/26/2022] [Indexed: 11/28/2022] Open
Abstract
Hypoxia is a common hallmark of healthy tissues in physiological states or chronically inflamed tissues in pathological states. Mammalian cells sense and adapt to hypoxia mainly through hypoxia-inducible factor (HIF) signaling. Many studies have shown that hypoxia and HIF signaling play an important regulatory role in development and function of innate immune cells and T cells, but their role in B cell biology is still controversial. B cells experience a complex life cycle (including hematopoietic stem cells, pro-B cells, pre-B cells, immature B cells, mature naïve B cells, activated B cells, plasma cells, and memory B cells), and the partial pressure of oxygen (PO2) in the corresponding developmental niche of stage-specific B cells is highly dynamic, which suggests that hypoxia and HIF signaling may play an indispensable role in B cell biology. Based on the fact that hypoxia niches exist in the B cell life cycle, this review focuses on recent discoveries about how hypoxia and HIF signaling regulate the development, metabolism, and function of B cells, to facilitate a deep understanding of the role of hypoxia in B cell-mediated adaptive immunity and to provide novel strategies for vaccine adjuvant research and the treatment of immunity-related or infectious diseases.
Collapse
Affiliation(s)
- Jinwei Zhang
- Chongqing Academy of Animal Sciences, Chongqing, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
- Chongqing Camab Biotech Ltd., Chongqing, China
| | - Xiaoqian Wu
- Chongqing Academy of Animal Sciences, Chongqing, China
| | - Jideng Ma
- Chongqing Academy of Animal Sciences, Chongqing, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Keren Long
- Chongqing Academy of Animal Sciences, Chongqing, China
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Jing Sun
- Chongqing Academy of Animal Sciences, Chongqing, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
| | - Mingzhou Li
- Farm Animal Genetic Resource Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
- Chongqing Camab Biotech Ltd., Chongqing, China
| |
Collapse
|
33
|
Dozzo A, Galvin A, Shin JW, Scalia S, O'Driscoll CM, Ryan KB. Modelling acute myeloid leukemia (AML): What's new? A transition from the classical to the modern. Drug Deliv Transl Res 2022:10.1007/s13346-022-01189-4. [PMID: 35930221 DOI: 10.1007/s13346-022-01189-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 11/24/2022]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous malignancy affecting myeloid cells in the bone marrow (BM) but can spread giving rise to impaired hematopoiesis. AML incidence increases with age and is associated with poor prognostic outcomes. There has been a disconnect between the success of novel drug compounds observed in preclinical studies of hematological malignancy and less than exceptional therapeutic responses in clinical trials. This review aims to provide a state-of-the-art overview on the different preclinical models of AML available to expand insights into disease pathology and as preclinical screening tools. Deciphering the complex physiological and pathological processes and developing predictive preclinical models are key to understanding disease progression and fundamental in the development and testing of new effective drug treatments. Standard scaffold-free suspension models fail to recapitulate the complex environment where AML occurs. To this end, we review advances in scaffold/matrix-based 3D models and outline the most recent advances in on-chip technology. We also provide an overview of clinically relevant animal models and review the expanding use of patient-derived samples, which offer the prospect to create more "patient specific" screening tools either in the guise of 3D matrix models, microphysiological "organ-on-chip" tools or xenograft models and discuss representative examples.
Collapse
Affiliation(s)
| | - Aoife Galvin
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Jae-Won Shin
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, 909 S. Wolcott Ave, Chicago, IL, 5091 COMRB, USA
| | - Santo Scalia
- Università degli Studi di Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Caitriona M O'Driscoll
- School of Pharmacy, University College Cork, Cork, Ireland.,SSPC Centre for Pharmaceutical Research, School of Pharmacy, University College Cork, Cork, Ireland
| | - Katie B Ryan
- School of Pharmacy, University College Cork, Cork, Ireland. .,SSPC Centre for Pharmaceutical Research, School of Pharmacy, University College Cork, Cork, Ireland.
| |
Collapse
|
34
|
Abstract
Hematopoietic stem cell (HSC) regeneration is the remarkable process by which extremely rare, normally inactive cells of the bone marrow can replace an entire organ if called to do so by injury or harnessed by transplantation. HSC research is arguably the first quantitative single-cell science and the foundation of adult stem cell biology. Bone marrow transplant is the oldest and most refined technique of regenerative medicine. Here we review the intertwined history of the discovery of HSCs and bone marrow transplant, the molecular and cellular mechanisms of HSC self-renewal, and the use of HSCs and their derivatives for cell therapy.
Collapse
Affiliation(s)
- Mitch Biermann
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Tannishtha Reya
- Department of Medicine, University of California San Diego, La Jolla, California 92093
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
35
|
Liver Regeneration by Hematopoietic Stem Cells: Have We Reached the End of the Road? Cells 2022; 11:cells11152312. [PMID: 35954155 PMCID: PMC9367594 DOI: 10.3390/cells11152312] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 02/01/2023] Open
Abstract
The liver is the organ with the highest regenerative capacity in the human body. However, various insults, including viral infections, alcohol or drug abuse, and metabolic overload, may cause chronic inflammation and fibrosis, leading to irreversible liver dysfunction. Despite advances in surgery and pharmacological treatments, liver diseases remain a leading cause of death worldwide. To address the shortage of donor liver organs for orthotopic liver transplantation, cell therapy in liver disease has emerged as a promising regenerative treatment. Sources include primary hepatocytes or functional hepatocytes generated from the reprogramming of induced pluripotent stem cells (iPSC). Different types of stem cells have also been employed for transplantation to trigger regeneration, including hematopoietic stem cells (HSCs), mesenchymal stromal cells (MSCs), endothelial progenitor cells (EPCs) as well as adult and fetal liver progenitor cells. HSCs, usually defined by the expression of CD34 and CD133, and MSCs, defined by the expression of CD105, CD73, and CD90, are attractive sources due to their autologous nature, ease of isolation and cryopreservation. The present review focuses on the use of bone marrow HSCs for liver regeneration, presenting evidence for an ongoing crosstalk between the hematopoietic and the hepatic system. This relationship commences during embryogenesis when the fetal liver emerges as the crossroads between the two systems converging the presence of different origins of cells (mesoderm and endoderm) in the same organ. Ample evidence indicates that the fetal liver supports the maturation and expansion of HSCs during development but also later on in life. Moreover, the fact that the adult liver remains one of the few sites for extramedullary hematopoiesis—albeit pathological—suggests that this relationship between the two systems is ongoing. Can, however, the hematopoietic system offer similar support to the liver? The majority of clinical studies using hematopoietic cell transplantation in patients with liver disease report favourable observations. The underlying mechanism—whether paracrine, fusion or transdifferentiation or a combination of the three—remains to be confirmed.
Collapse
|
36
|
Li M, Morse B, Kassim S. Development and clinical translation considerations for the next wave of gene modified hematopoietic stem and progenitor cells therapies. Expert Opin Biol Ther 2022; 22:1177-1191. [PMID: 35833356 DOI: 10.1080/14712598.2022.2101361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Consistent and reliable manufacture of gene modified hematopoietic stem and progenitor cell (HPSC) therapies will be of the utmost importance as they become more mainstream and address larger populations. Robust development campaigns will be needed to ensure that these products will be delivered to patients with the highest quality standards. AREAS COVERED Through publicly available manuscripts, press releases, and news articles - this review touches on aspects related to HSPC therapy, development, and manufacturing. EXPERT OPINION Recent advances in genome modification technology coupled with the longstanding clinical success of HSPCs warrants great optimism for the next generation of engineered HSPC-based therapies. Treatments for some diseases that have thus far been intractable now appear within reach. Reproducible manufacturing will be of critical importance in delivering these therapies but will be challenging due to the need for bespoke materials and methods in combination with the lack of off-the-shelf solutions. Continued progress in the field will manifest in the form of industrialization which currently requires attention and resources directed toward the custom reagents, a focus on closed and automated processes, and safer and more precise genome modification technologies that will enable broader, faster, and safer access to these life-changing therapies.
Collapse
Affiliation(s)
| | - Brent Morse
- Dark Horse Consulting Group, Walnut Creek, CA, USA
| | | |
Collapse
|
37
|
Garcia-Gerique L, García M, Garrido-Garcia A, Gómez-González S, Torrebadell M, Prada E, Pascual-Pasto G, Muñoz O, Perez-Jaume S, Lemos I, Salvador N, Vila-Ubach M, Doncel-Requena A, Suñol M, Carcaboso AM, Mora J, Lavarino C. MIF/CXCR4 signaling axis contributes to survival, invasion, and drug resistance of metastatic neuroblastoma cells in the bone marrow microenvironment. BMC Cancer 2022; 22:669. [PMID: 35715791 PMCID: PMC9206243 DOI: 10.1186/s12885-022-09725-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 05/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The bone marrow (BM) is the most common site of dissemination in patients with aggressive, metastatic neuroblastoma (NB). However, the molecular mechanisms underlying the aggressive behavior of NB cells in the BM niche are still greatly unknown. In the present study, we explored biological mechanisms that play a critical role in NB cell survival and progression in the BM and investigated potential therapeutic targets. METHODS Patient-derived bone marrow (BM) primary cultures were generated using fresh BM aspirates obtained from NB patients. NB cell lines were cultured in the presence of BM conditioned media containing cell-secreted factors, and under low oxygen levels (1% O2) to mimic specific features of the BM microenvironment of high-risk NB patients. The BM niche was explored using cytokine profiling assays, cell migration-invasion and viability assays, flow cytometry and analysis of RNA-sequencing data. Selective pharmacological inhibition of factors identified as potential mediators of NB progression within the BM niche was performed in vitro and in vivo. RESULTS We identified macrophage migration inhibitory factor (MIF) as a key inflammatory cytokine involved in BM infiltration. Cytokine profiling and RNA-sequencing data analysis revealed NB cells as the main source of MIF in the BM, suggesting a potential role of MIF in tumor invasion. Exposure of NB cells to BM-conditions increased NB cell-surface expression of the MIF receptor CXCR4, which was associated with increased cell viability, enhanced migration-invasion, and activation of PI3K/AKT and MAPK/ERK signaling pathways. Moreover, subcutaneous co-injection of NB and BM cells enhanced tumor engraftment in mice. MIF inhibition with 4-IPP impaired in vitro NB aggressiveness, and improved drug response while delayed NB growth, improving survival of the NB xenograft model. CONCLUSIONS Our findings suggest that BM infiltration by NB cells may be mediated, in part, by MIF-CXCR4 signaling. We demonstrate the antitumor efficacy of MIF targeting in vitro and in vivo that could represent a novel therapeutic target for patients with disseminated high-risk NB.
Collapse
Affiliation(s)
- Laura Garcia-Gerique
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Marta García
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Alícia Garrido-Garcia
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Soledad Gómez-González
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Montserrat Torrebadell
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Estela Prada
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Guillem Pascual-Pasto
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Oscar Muñoz
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.,Department of Pathology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Sara Perez-Jaume
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Isadora Lemos
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Noelia Salvador
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Monica Vila-Ubach
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Ana Doncel-Requena
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Mariona Suñol
- Department of Pathology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Angel M Carcaboso
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Jaume Mora
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.,Laboratory of Molecular Oncology, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu, Barcelona, Spain
| | - Cinzia Lavarino
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain. .,Laboratory of Molecular Oncology, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu, Barcelona, Spain.
| |
Collapse
|
38
|
Gudmundsson KO, Du Y. Quiescence regulation by normal haematopoietic stem cells and leukaemia stem cells. FEBS J 2022. [PMID: 35514133 DOI: 10.1111/febs.16472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 03/21/2022] [Accepted: 05/03/2022] [Indexed: 11/30/2022]
Abstract
The haematopoietic system is maintained by rare haematopoietic stem cells (HSCs), which are quiescent most of the time and only divide occasionally to self-renew and/or to undergo commitment to clonal expansion via the generation of highly proliferative progenitor cells. The latter is responsible for the generation of all mature cells of the system through subsequent lineage commitment and terminal differentiation. Cells with similar properties also exist in leukaemias and are known as leukaemia stem cells (LSCs). Quiescence provides essential protection for both HSC and LSC from cytotoxic stress and DNA damage and, in the case of LSCs, likely causes therapy resistance and disease relapse in leukaemia patients. Specific inhibition of LSC quiescence has been considered a promising strategy for eliminating LSCs and curing leukaemias. Although the understanding of mechanisms responsible for quiescence maintenance in these cells remains limited, particularly for LSCs, recent studies have suggested potential differences in their dependency on certain pathways and their levels of stress and DNA damage caused by increased cycling. Such differences likely stem from oncogenic mutations in LSCs and could be specifically exploited for the elimination of LSCs while sparing normal HSCs in the future.
Collapse
Affiliation(s)
- Kristbjorn Orri Gudmundsson
- Basic Science Program Leidos Biomedical Research Inc. Frederick National Laboratory for Cancer Research in the Mouse Cancer Genetics Program Center for Cancer Research NCI Frederick MD USA
| | - Yang Du
- Department of Pediatrics Uniformed Services University of the Health Sciences Bethesda MD USA
| |
Collapse
|
39
|
Current insights into the bone marrow niche: From biology in vivo to bioengineering ex vivo. Biomaterials 2022; 286:121568. [DOI: 10.1016/j.biomaterials.2022.121568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 11/21/2022]
|
40
|
Hypoxia orchestrates the lymphovascular–immune ensemble in cancer. Trends Cancer 2022; 8:771-784. [DOI: 10.1016/j.trecan.2022.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/31/2022] [Accepted: 04/25/2022] [Indexed: 11/18/2022]
|
41
|
Shin DY. Human acute myeloid leukemia stem cells: evolution of concept. Blood Res 2022; 57:67-74. [PMID: 35483929 PMCID: PMC9057671 DOI: 10.5045/br.2022.2021221] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/23/2022] [Accepted: 03/25/2022] [Indexed: 11/17/2022] Open
Abstract
The history of human acute myeloid leukemia stem cells (AMLSCs) began in a seminal study performed by Lapidot and Dick, proving that only CD34+CD38- human primary acute myeloid leukemia (AML) cells can repopulate in severe combined immunodeficient mice. The concept of leukemic stem cells (LSCs) has impeded a huge change in the treatment strategy against AML from killing proliferating leukemic cells to eradicating quiescent/dormant LSCs. As next-generation sequencing technologies have developed, multiple and recurrent genetic mutations have been discovered in large cohorts of patients with AML, and the updated understanding of leukemogenesis has improved the old concept of LSC to a revised version of a serial developmental model of LSC; that is, pre-LSCs are generated as seeds by the first hit on epigenetic regulators, and then, leukemia-initiating LSCs emerge from seeds by the second hits on genes involved in transcription and signaling. Dreams for universal and targetable AMLSC biomarker sparing healthy hematopoietic stem cells have weakened after the confrontation of significant heterogeneity of AMLSCs from genomic and immunophenotypic viewpoints. However, there is still hope for effective targets for AMLSCs since there is evidence that grouped gene signatures, such as 17-gene LSC score, and common epigenetic signatures, such as HOXA clusters, independent of various gene mutations, exist. Recently, the LSC niche in the bone marrow has been actively investigated and has expanded our knowledge of the physiology and vulnerability of AMLSCs. Presently, an applicable treatment that always works in AMLSCs is lacking. However, we will find a way, we always have.
Collapse
Affiliation(s)
- Dong-Yeop Shin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
42
|
Kandarakov O, Belyavsky A, Semenova E. Bone Marrow Niches of Hematopoietic Stem and Progenitor Cells. Int J Mol Sci 2022; 23:ijms23084462. [PMID: 35457280 PMCID: PMC9032554 DOI: 10.3390/ijms23084462] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/15/2022] Open
Abstract
The mammalian hematopoietic system is remarkably efficient in meeting an organism’s vital needs, yet is highly sensitive and exquisitely regulated. Much of the organismal control over hematopoiesis comes from the regulation of hematopoietic stem cells (HSCs) by specific microenvironments called niches in bone marrow (BM), where HSCs reside. The experimental studies of the last two decades using the most sophisticated and advanced techniques have provided important data on the identity of the niche cells controlling HSCs functions and some mechanisms underlying niche-HSC interactions. In this review we discuss various aspects of organization and functioning of the HSC cell niche in bone marrow. In particular, we review the anatomy of BM niches, various cell types composing the niche, niches for more differentiated cells, metabolism of HSCs in relation to the niche, niche aging, leukemic transformation of the niche, and the current state of HSC niche modeling in vitro.
Collapse
|
43
|
Chicana B, Abbasizadeh N, Burns C, Taglinao H, Spencer JA, Manilay JO. Deletion of Vhl in Dmp1-Expressing Cells Causes Microenvironmental Impairment of B Cell Lymphopoiesis. Front Immunol 2022; 13:780945. [PMID: 35250971 PMCID: PMC8889104 DOI: 10.3389/fimmu.2022.780945] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/11/2022] [Indexed: 12/25/2022] Open
Abstract
The contributions of skeletal cells to the processes of B cell development in the bone marrow (BM) have not been completely described. The von-Hippel Lindau protein (VHL) plays a key role in cellular responses to hypoxia. Previous work showed that Dmp1-Cre;Vhl conditional knockout mice (VhlcKO), which deletes Vhl in subsets of mesenchymal stem cells, late osteoblasts and osteocytes, display dysregulated bone growth and reduction in B cells. Here, we investigated the mechanisms underlying the B cell defects using flow cytometry and high-resolution imaging. In the VhlcKO BM, B cell progenitors were increased in frequency and number, whereas Hardy Fractions B-F were decreased. VhlcKO Fractions B-C cells showed increased apoptosis and quiescence. Reciprocal BM chimeras confirmed a B cell-extrinsic source of the VhlcKO B cell defects. In support of this, VhlcKO BM supernatant contained reduced CXCL12 and elevated EPO levels. Intravital and ex vivo imaging revealed VhlcKO BM blood vessels with increased diameter, volume, and a diminished blood-BM barrier. Staining of VhlcKO B cells with an intracellular hypoxic marker indicated the natural existence of distinct B cell microenvironments that differ in local oxygen tensions and that the B cell developmental defects in VhlcKO BM are not initiated by hypoxia. Our studies identify novel mechanisms linking altered bone homeostasis with drastic BM microenvironmental changes that dysregulate B cell development.
Collapse
Affiliation(s)
- Betsabel Chicana
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Nastaran Abbasizadeh
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States.,Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, United States
| | - Christian Burns
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States.,Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, United States
| | - Hanna Taglinao
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| | - Joel A Spencer
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States.,Department of Bioengineering, School of Engineering, University of California, Merced, Merced, CA, United States.,Bioengineering Graduate Program, University of California, Merced, Merced, CA, United States
| | - Jennifer O Manilay
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States.,Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| |
Collapse
|
44
|
Mayer IM, Hoelbl-Kovacic A, Sexl V, Doma E. Isolation, Maintenance and Expansion of Adult Hematopoietic Stem/Progenitor Cells and Leukemic Stem Cells. Cancers (Basel) 2022; 14:1723. [PMID: 35406494 PMCID: PMC8996967 DOI: 10.3390/cancers14071723] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 12/12/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are rare, self-renewing cells that perch on top of the hematopoietic tree. The HSCs ensure the constant supply of mature blood cells in a tightly regulated process producing peripheral blood cells. Intense efforts are ongoing to optimize HSC engraftment as therapeutic strategy to treat patients suffering from hematopoietic diseases. Preclinical research paves the way by developing methods to maintain, manipulate and expand HSCs ex vivo to understand their regulation and molecular make-up. The generation of a sufficient number of transplantable HSCs is the Holy Grail for clinical therapy. Leukemia stem cells (LSCs) are characterized by their acquired stem cell characteristics and are responsible for disease initiation, progression, and relapse. We summarize efforts, that have been undertaken to increase the number of long-term (LT)-HSCs and to prevent differentiation towards committed progenitors in ex vivo culture. We provide an overview and compare methods currently available to isolate, maintain and enrich HSC subsets, progenitors and LSCs and discuss their individual advantages and drawbacks.
Collapse
Affiliation(s)
| | | | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (I.M.M.); (A.H.-K.); (E.D.)
| | | |
Collapse
|
45
|
Pircher T, Wackerhage H, Akova E, Böcker W, Aszodi A, Saller MM. Fusion of Normoxic- and Hypoxic-Preconditioned Myoblasts Leads to Increased Hypertrophy. Cells 2022; 11:cells11061059. [PMID: 35326510 PMCID: PMC8947054 DOI: 10.3390/cells11061059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/14/2022] [Accepted: 03/19/2022] [Indexed: 12/20/2022] Open
Abstract
Injuries, high altitude, and endurance exercise lead to hypoxic conditions in skeletal muscle and sometimes to hypoxia-induced local tissue damage. Thus, regenerative myoblasts/satellite cells are exposed to different levels and durations of partial oxygen pressure depending on the spatial distance from the blood vessels. To date, it is unclear how hypoxia affects myoblasts proliferation, differentiation, and particularly fusion with normoxic myoblasts. To study this, we investigated how 21% and 2% oxygen affects C2C12 myoblast morphology, proliferation, and myogenic differentiation and evaluated the fusion of normoxic- or hypoxic-preconditioned C2C12 cells in 21% or 2% oxygen in vitro. Out data show that the long-term hypoxic culture condition does not affect the proliferation of C2C12 cells but leads to rounder cells and reduced myotube formation when compared with myoblasts exposed to normoxia. However, when normoxic- and hypoxic-preconditioned myoblasts were differentiated together, the resultant myotubes were significantly larger than the control myotubes. Whole transcriptome sequencing analysis revealed several novel candidate genes that are differentially regulated during the differentiation under normoxia and hypoxia in mixed culture conditions and may thus be involved in the increase in myotube size. Taken together, oxygen-dependent adaption and interaction of myoblasts may represent a novel approach for the development of innovative therapeutic targets.
Collapse
Affiliation(s)
- Tamara Pircher
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (T.P.); (E.A.); (W.B.); (A.A.)
| | - Henning Wackerhage
- Faculty of Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60, 80992 Munich, Germany;
| | - Elif Akova
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (T.P.); (E.A.); (W.B.); (A.A.)
| | - Wolfgang Böcker
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (T.P.); (E.A.); (W.B.); (A.A.)
| | - Attila Aszodi
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (T.P.); (E.A.); (W.B.); (A.A.)
| | - Maximilian M. Saller
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (T.P.); (E.A.); (W.B.); (A.A.)
- Correspondence: ; Tel.: +49-89-4400-55486
| |
Collapse
|
46
|
Caballano-Infantes E, Cahuana GM, Bedoya FJ, Salguero-Aranda C, Tejedo JR. The Role of Nitric Oxide in Stem Cell Biology. Antioxidants (Basel) 2022; 11:497. [PMID: 35326146 PMCID: PMC8944807 DOI: 10.3390/antiox11030497] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a gaseous biomolecule endogenously synthesized with an essential role in embryonic development and several physiological functions, such as regulating mitochondrial respiration and modulation of the immune response. The dual role of NO in embryonic stem cells (ESCs) has been previously reported, preserving pluripotency and cell survival or inducing differentiation with a dose-dependent pattern. In this line, high doses of NO have been used in vitro cultures to induce focused differentiation toward different cell lineages being a key molecule in the regenerative medicine field. Moreover, optimal conditions to promote pluripotency in vitro are essential for their use in advanced therapies. In this sense, the molecular mechanisms underlying stemness regulation by NO have been studied intensively over the current years. Recently, we have reported the role of low NO as a hypoxia-like inducer in pluripotent stem cells (PSCs), which supports using this molecule to maintain pluripotency under normoxic conditions. In this review, we stress the role of NO levels on stem cells (SCs) fate as a new approach for potential cell therapy strategies. Furthermore, we highlight the recent uses of NO in regenerative medicine due to their properties regulating SCs biology.
Collapse
Affiliation(s)
- Estefanía Caballano-Infantes
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain
| | - Gladys Margot Cahuana
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco Javier Bedoya
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carmen Salguero-Aranda
- Department of Pathology, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, CSIC-University of Seville, 41013 Seville, Spain;
- Spanish Biomedical Research Network Centre in Oncology-CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41004 Seville, Spain
| | - Juan R. Tejedo
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
47
|
Abstract
Metabolism has been studied mainly in cultured cells or at the level of whole tissues or whole organisms in vivo. Consequently, our understanding of metabolic heterogeneity among cells within tissues is limited, particularly when it comes to rare cells with biologically distinct properties, such as stem cells. Stem cell function, tissue regeneration and cancer suppression are all metabolically regulated, although it is not yet clear whether there are metabolic mechanisms unique to stem cells that regulate their activity and function. Recent work has, however, provided evidence that stem cells do have a metabolic signature that is distinct from that of restricted progenitors and that metabolic changes influence tissue homeostasis and regeneration. Stem cell maintenance throughout life in many tissues depends upon minimizing anabolic pathway activation and cell division. Consequently, stem cell activation by tissue injury is associated with changes in mitochondrial function, lysosome activity and lipid metabolism, potentially at the cost of eroding self-renewal potential. Stem cell metabolism is also regulated by the environment: stem cells metabolically interact with other cells in their niches and are able to sense and adapt to dietary changes. The accelerating understanding of stem cell metabolism is revealing new aspects of tissue homeostasis with the potential to promote tissue regeneration and cancer suppression.
Collapse
|
48
|
Park JH, Lee HK. Current Understanding of Hypoxia in Glioblastoma Multiforme and Its Response to Immunotherapy. Cancers (Basel) 2022; 14:1176. [PMID: 35267480 PMCID: PMC8909860 DOI: 10.3390/cancers14051176] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/20/2022] [Accepted: 02/23/2022] [Indexed: 02/06/2023] Open
Abstract
Hypoxia is a hallmark of glioblastoma multiforme (GBM), the most aggressive cancer of the central nervous system, and is associated with multiple aspects of tumor pathogenesis. For example, hypoxia induces resistance to conventional cancer therapies and inhibits antitumor immune responses. Thus, targeting hypoxia is an attractive strategy for GBM therapy. However, traditional studies on hypoxia have largely excluded the immune system. Recently, the critical role of the immune system in the defense against multiple tumors has become apparent, leading to the development of effective immunotherapies targeting numerous cancer types. Critically, however, GBM is classified as a "cold tumor" due to poor immune responses. Thus, to improve GBM responsiveness against immunotherapies, an improved understanding of both immune function in GBM and the role of hypoxia in mediating immune responses within the GBM microenvironment is needed. In this review, we discuss the role of hypoxia in GBM from a clinical, pathological, and immunological perspective.
Collapse
Affiliation(s)
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea;
| |
Collapse
|
49
|
Janbandhu V, Tallapragada V, Patrick R, Li Y, Abeygunawardena D, Humphreys DT, Martin EM, Ward AO, Contreras O, Farbehi N, Yao E, Du J, Dunwoodie SL, Bursac N, Harvey RP. Hif-1a suppresses ROS-induced proliferation of cardiac fibroblasts following myocardial infarction. Cell Stem Cell 2022; 29:281-297.e12. [PMID: 34762860 PMCID: PMC9021927 DOI: 10.1016/j.stem.2021.10.009] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 06/16/2021] [Accepted: 10/20/2021] [Indexed: 02/07/2023]
Abstract
We report that cardiac fibroblasts (CFs) and mesenchymal progenitors are more hypoxic than other cardiac interstitial populations, express more hypoxia-inducible factor 1α (HIF-1α), and exhibit increased glycolytic metabolism. CF-specific deletion of Hif-1a resulted in decreased HIF-1 target gene expression and increased mesenchymal progenitors in uninjured hearts and increased CF activation without proliferation following sham injury, as demonstrated using single-cell RNA sequencing (scRNA-seq). After myocardial infarction (MI), however, there was ∼50% increased CF proliferation and excessive scarring and contractile dysfunction, a scenario replicated in 3D engineered cardiac microtissues. CF proliferation was associated with higher reactive oxygen species (ROS) as occurred also in wild-type mice treated with the mitochondrial ROS generator MitoParaquat (MitoPQ). The mitochondrial-targeted antioxidant MitoTEMPO rescued Hif-1a mutant phenotypes. Thus, HIF-1α in CFs provides a critical braking mechanism against excessive post-ischemic CF activation and proliferation through regulation of mitochondrial ROS. CFs are potential cellular targets for designer antioxidant therapies in cardiovascular disease.
Collapse
Affiliation(s)
- Vaibhao Janbandhu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia,St. Vincent’s Clinical School, UNSW Sydney, NSW, Australia,Correspondence: (V.J.), (R.P.H.)
| | - Vikram Tallapragada
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia,St. Vincent’s Clinical School, UNSW Sydney, NSW, Australia
| | - Ralph Patrick
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia,St. Vincent’s Clinical School, UNSW Sydney, NSW, Australia
| | - Yanzhen Li
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Dhanushi Abeygunawardena
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia,School of Biotechnology and Biomolecular Science, University of New South Wales, Sydney, NSW, Australia
| | - David T. Humphreys
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia,St. Vincent’s Clinical School, UNSW Sydney, NSW, Australia
| | | | - Alexander O. Ward
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia,St. Vincent’s Clinical School, UNSW Sydney, NSW, Australia
| | - Osvaldo Contreras
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia,St. Vincent’s Clinical School, UNSW Sydney, NSW, Australia
| | - Nona Farbehi
- Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research Sydney, NSW 2010, Australia
| | - Ernestene Yao
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Junjie Du
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Sally L. Dunwoodie
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia,St. Vincent’s Clinical School, UNSW Sydney, NSW, Australia
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA,Department of Medicine, Duke University, Durham, NC 27708, USA
| | - Richard P. Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia,St. Vincent’s Clinical School, UNSW Sydney, NSW, Australia,School of Biotechnology and Biomolecular Science, University of New South Wales, Sydney, NSW, Australia,Lead contact,Correspondence: (V.J.), (R.P.H.)
| |
Collapse
|
50
|
Zhao Y, He J, Zhu T, Zhang Y, Zhai Y, Xue P, Yao Y, Zhou Z, He M, Qu W, Zhang Y. Cadmium exposure reprograms energy metabolism of hematopoietic stem cells to promote myelopoiesis at the expense of lymphopoiesis in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 231:113208. [PMID: 35051759 DOI: 10.1016/j.ecoenv.2022.113208] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/08/2022] [Accepted: 01/13/2022] [Indexed: 06/14/2023]
Abstract
Cadmium (Cd) is a highly toxic heavy metal in our living environment. Hematopoietic stem cells (HSC) are ancestors for all blood cells. Therefore understanding the impact of Cd on HSC is significant for public health. The aim of this study was to investigate the impact of Cd2+ on energy metabolism of HSC and its involvement in hematopoiesis. Wild-type C57BL/6 mice were treated with 10 ppm of Cd2+ via drinking water for 3 months, and thereafter glycolysis and mitochondrial (MT) oxidative phosphorylation (OXPHOS) of HSC in the bone marrow (BM) and their impact on hematopoiesis were evaluated. After Cd2+ treatment, HSC had reduced lactate dehydrogenase (LDH) activity and lactate production while having increased pyruvate dehydrogenase (PDH) activity, MT membrane potential, ATP production, oxygen (O2) consumption and reactive oxygen species (ROS), indicating that Cd2+ switched the pattern of energy metabolism from glycolysis to OXPHOS in HSC. Moreover, Cd2+ switch of HSC energy metabolism was critically dependent on Wnt5a/Cdc42/calcium (Ca2+) signaling triggered by a direct action of Cd2+ on HSC. To test the biological significance of Cd2+ impact on HSC energy metabolism, HSC were intervened for Ca2+, OXPHOS, or ROS in vitro, and thereafter the HSC were transplanted into lethally irradiated recipients to reconstitute the immune system; the transplantation assay indicated that Ca2+-dependent MT OXPHOS dominated the skewed myelopoiesis of HSC by Cd2+ exposure. Collectively, we revealed that Cd2+ exposure activated Wnt5a/Cdc42/Ca2+ signaling to reprogram the energy metabolism of HSC to drive myelopoiesis at the expense of lymphopoiesis.
Collapse
Affiliation(s)
- Yifan Zhao
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Jinyi He
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Tingting Zhu
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Yufan Zhang
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Yue Zhai
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Peng Xue
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Ye Yao
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Zhijun Zhou
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Miao He
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Sciences, Fudan University, Shanghai 200032, China
| | - Weidong Qu
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China
| | - Yubin Zhang
- School of Public Health and Key Laboratory of Public Health Safety, MOE, Fudan University, Shanghai 200032, China.
| |
Collapse
|