101
|
p21-Activated kinases as promising therapeutic targets in hematological malignancies. Leukemia 2022; 36:315-326. [PMID: 34697424 DOI: 10.1038/s41375-021-01451-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 01/12/2023]
Abstract
The p21-Activated Kinases (PAKs) are a family of six serine/threonine kinases that were originally identified as downstream effectors of the Rho GTPases Cdc42 and Rac. Since the first PAK was discovered in 1994, studies have revealed their fundamental and biological importance in the development of physiological systems. Within the cell, PAKs also play significant roles in regulating essential cellular processes such as cytoskeletal dynamics, gene expression, cell survival, and cell cycle progression. These processes are often deregulated in numerous cancers when different PAKs are overexpressed or amplified at the chromosomal level. Furthermore, PAKs modulate multiple oncogenic signaling pathways which facilitate apoptosis escape, uncontrolled proliferation, and drug resistance. There is growing insight into the critical roles of PAKs in regulating steady-state hematopoiesis, including the properties of hematopoietic stem cells (HSC), and the initiation and progression of hematological malignancies. This review will focus on the most recent studies that provide experimental evidence showing how specific PAKs regulate the properties of leukemic stem cells (LSCs) and drug-resistant cells to initiate and maintain hematological malignancies. The current understanding of the molecular and cellular mechanisms by which the PAKs operate in specific human leukemia or lymphomas will be discussed. From a translational point of view, PAKs have been suggested to be critical therapeutic targets and potential prognosis markers; thus, this review will also discuss current therapeutic strategies against hematological malignancies using existing small-molecule PAK inhibitors, as well as promising combination treatments, to sensitize drug-resistant cells to conventional therapies. The challenges of toxicity and non-specific targeting associated with some PAK inhibitors, as well as how future approaches for PAK inhibition to overcome these limitations, will also be addressed.
Collapse
|
102
|
Rinkevich B, Ballarin L, Martinez P, Somorjai I, Ben‐Hamo O, Borisenko I, Berezikov E, Ereskovsky A, Gazave E, Khnykin D, Manni L, Petukhova O, Rosner A, Röttinger E, Spagnuolo A, Sugni M, Tiozzo S, Hobmayer B. A pan-metazoan concept for adult stem cells: the wobbling Penrose landscape. Biol Rev Camb Philos Soc 2022; 97:299-325. [PMID: 34617397 PMCID: PMC9292022 DOI: 10.1111/brv.12801] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022]
Abstract
Adult stem cells (ASCs) in vertebrates and model invertebrates (e.g. Drosophila melanogaster) are typically long-lived, lineage-restricted, clonogenic and quiescent cells with somatic descendants and tissue/organ-restricted activities. Such ASCs are mostly rare, morphologically undifferentiated, and undergo asymmetric cell division. Characterized by 'stemness' gene expression, they can regulate tissue/organ homeostasis, repair and regeneration. By contrast, analysis of other animal phyla shows that ASCs emerge at different life stages, present both differentiated and undifferentiated phenotypes, and may possess amoeboid movement. Usually pluri/totipotent, they may express germ-cell markers, but often lack germ-line sequestering, and typically do not reside in discrete niches. ASCs may constitute up to 40% of animal cells, and participate in a range of biological phenomena, from whole-body regeneration, dormancy, and agametic asexual reproduction, to indeterminate growth. They are considered legitimate units of selection. Conceptualizing this divergence, we present an alternative stemness metaphor to the Waddington landscape: the 'wobbling Penrose' landscape. Here, totipotent ASCs adopt ascending/descending courses of an 'Escherian stairwell', in a lifelong totipotency pathway. ASCs may also travel along lower stemness echelons to reach fully differentiated states. However, from any starting state, cells can change their stemness status, underscoring their dynamic cellular potencies. Thus, vertebrate ASCs may reflect just one metazoan ASC archetype.
Collapse
Affiliation(s)
- Baruch Rinkevich
- Israel Oceanographic & Limnological ResearchNational Institute of OceanographyPOB 9753, Tel ShikmonaHaifa3109701Israel
| | - Loriano Ballarin
- Department of BiologyUniversity of PadovaVia Ugo Bassi 58/BPadova35121Italy
| | - Pedro Martinez
- Departament de Genètica, Microbiologia i EstadísticaUniversitat de BarcelonaAv. Diagonal 643Barcelona08028Spain
- Institut Català de Recerca i Estudis Avançats (ICREA)Passeig Lluís Companys 23Barcelona08010Spain
| | - Ildiko Somorjai
- School of BiologyUniversity of St AndrewsSt Andrews, FifeKY16 9ST, ScotlandUK
| | - Oshrat Ben‐Hamo
- Israel Oceanographic & Limnological ResearchNational Institute of OceanographyPOB 9753, Tel ShikmonaHaifa3109701Israel
| | - Ilya Borisenko
- Department of Embryology, Faculty of BiologySaint‐Petersburg State UniversityUniversity Embankment, 7/9Saint‐Petersburg199034Russia
| | - Eugene Berezikov
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center GroningenAntonius Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Alexander Ereskovsky
- Department of Embryology, Faculty of BiologySaint‐Petersburg State UniversityUniversity Embankment, 7/9Saint‐Petersburg199034Russia
- Institut Méditerranéen de Biodiversité et d'Ecologie marine et continentale (IMBE), Aix Marseille University, CNRS, IRD, Avignon UniversityJardin du Pharo, 58 Boulevard Charles LivonMarseille13007France
- Koltzov Institute of Developmental Biology of Russian Academy of SciencesUlitsa Vavilova, 26Moscow119334Russia
| | - Eve Gazave
- Université de Paris, CNRS, Institut Jacques MonodParisF‐75006France
| | - Denis Khnykin
- Department of PathologyOslo University HospitalBygg 19, Gaustad Sykehus, Sognsvannsveien 21Oslo0188Norway
| | - Lucia Manni
- Department of BiologyUniversity of PadovaVia Ugo Bassi 58/BPadova35121Italy
| | - Olga Petukhova
- Collection of Vertebrate Cell CulturesInstitute of Cytology, Russian Academy of SciencesTikhoretsky Ave. 4St. Petersburg194064Russia
| | - Amalia Rosner
- Israel Oceanographic & Limnological ResearchNational Institute of OceanographyPOB 9753, Tel ShikmonaHaifa3109701Israel
| | - Eric Röttinger
- Université Côte d'Azur, CNRS, INSERM, Institute for Research on Cancer and Aging, Nice (IRCAN)Nice06107France
- Université Côte d'Azur, Federative Research Institute – Marine Resources (IFR MARRES)28 Avenue de ValroseNice06103France
| | - Antonietta Spagnuolo
- Department of Biology and Evolution of Marine OrganismsStazione Zoologica Anton DohrnVilla ComunaleNaples80121Italy
| | - Michela Sugni
- Department of Environmental Science and Policy (ESP)Università degli Studi di MilanoVia Celoria 26Milan20133Italy
| | - Stefano Tiozzo
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement de Villefranche‐sur‐mer (LBDV)06234 Villefranche‐sur‐MerVillefranche sur MerCedexFrance
| | - Bert Hobmayer
- Institute of Zoology and Center for Molecular Biosciences, University of InnsbruckTechnikerstrInnsbruck256020Austria
| |
Collapse
|
103
|
Zhu B, Zhong W, Cao X, Pan G, Xu M, Zheng J, Chen H, Feng X, Luo C, Lu C, Xiao J, Lin W, Lai C, Li M, Du X, Yi Q, Yan D. Loss of miR-31-5p drives hematopoietic stem cell malignant transformation and restoration eliminates leukemia stem cells in mice. Sci Transl Med 2022; 14:eabh2548. [PMID: 35080912 DOI: 10.1126/scitranslmed.abh2548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Leukemia stem cells (LSCs) propagate leukemia and are responsible for the high frequency of relapse of treated patients. The ability to target LSCs remains elusive, indicating a need to understand the underlying mechanism of LSC formation. Here, we report that miR-31-5p is reduced or undetectable in human LSCs compared to hematopoietic stem progenitor cells (HSPCs). Inhibition of miR-31-5p in HSPCs promotes the expression of its target gene FIH, encoding FIH [factor inhibiting hypoxia-inducing factor 1α (HIF-1α)], to suppress HIF-1α signaling. Increased FIH resulted in a switch from glycolysis to oxidative phosphorylation (OXPHOS) as the predominant mode of energy metabolism and increased the abundance of the oncometabolite fumarate. Increased fumarate promoted the conversion of HSPCs to LSCs and initiated myeloid leukemia-like disease in NOD-Prkdcscid IL2rgtm1/Bcgen (B-NDG) mice. We further demonstrated that miR-31-5p inhibited long- and short-term hematopoietic stem cells with a high frequency of LSCs. In combination with the chemotherapeutic agent Ara-C (cytosine arabinoside), restoration of miR-31-5p using G7 poly (amidoamine) nanosized dendriplex encapsulating miR-31-5p eliminated LSCs and inhibited acute myeloid leukemia (AML) progression in patient-derived xenograft mouse models. These results demonstrated a mechanism of HSC malignant transformation through altered energy metabolism and provided a potential therapeutic strategy to treat patients with AML.
Collapse
Affiliation(s)
- Biying Zhu
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Wenbin Zhong
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Xiuye Cao
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Guoping Pan
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Mengyang Xu
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Jie Zheng
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Huanzhao Chen
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Xiaoqin Feng
- Hematology and Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chengwei Luo
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510000, China
| | - Chen Lu
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Jie Xiao
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Weize Lin
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Chaofeng Lai
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Mingchuan Li
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| | - Xin Du
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510000, China
| | - Qing Yi
- Cancer Center, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Daoguang Yan
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
104
|
Moghnieh R, Khalil A, Bizri N, Francis N, Imad S, Mezher M, Mrad Z, Ibrahim J, Zahran K, Farroukh F, Itani M, Assaad A, Sinno L, Abdallah D, Ibrahim A. QTc prolongation during levofloxacin and triazole combination chemoprophylaxis: Prevalence and predisposing risk factors in a cohort of hematopoietic cell transplantation recipients. J Oncol Pharm Pract 2022; 29:534-542. [PMID: 35075933 DOI: 10.1177/10781552221074016] [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: 11/15/2022]
Abstract
BACKGROUND QTc interval prolongation has been reported when combining fluoroquinolones and triazoles for chemoprophylaxis in cancer patients. Herein, we aimed to identify the prevalence and contributing factors to QTc prolongation in hematopoietic cell transplantation (HCT) recipients who received these agents during the neutropenic phase. METHODS This is a retrospective medical chart review conducted at a university hospital in Lebanon from 2017 to 2020. It included all adult HCT inpatients on antimicrobial prophylaxis with fluoroquinolones and triazoles and whose baseline ECG monitoring done prior to chemoprophylaxis administration, then on day-3 and day-6 of therapy, were available. RESULTS Overall, 68 HCT recipients met our inclusion criteria, of which 22% developed QTc prolongation. Based on bivariate analysis, female gender contributed to QTc prolongation (P = 0.001). There was a trend to QTc prolongation in patients with predisposing thyroid disease (P = 0.12), grade 2 vomiting and diarrhea (P = 0.16, P = 0.46, respectively), baseline hypokalemia (P = 0.18) and hypocalcemia (P = 0.3), hypomagnesemia on day-3 (P = 0.21) and day-6 hyponatremia (P = 0.36). Patients receiving two or more drugs with a known or probable risk of QTc prolongation (other than the fluoroquinolone/ triazole combination) were more prone to experience a prolonged QTc interval (P = 0.09). None of the patients that had QTc prolongation died or developed serious arrhythmias. CONCLUSION The prevalence of QTc prolongation was 22% among HCT recipients on fluoroquinolone and triazole prophylaxis, yet we did not identify any independent risk factors for this issue. None of the patients that had QTc interval prolongation died or developed serious arrhythmias.
Collapse
Affiliation(s)
- Rima Moghnieh
- Department of Internal Medicine, Division of Infectious Diseases, Middle East Institute of Health University Hospital, Bsalim, Lebanon.,Department of Internal Medicine, Division of Infectious Diseases, 36696Makassed General Hospital, Beirut, Lebanon.,Faculty of Medicine, Lebanese University, Beirut, Lebanon
| | - Ahmad Khalil
- Department of Internal Medicine, Division of Hematology, Oncology and Bone Marrow Transplantation, Middle East Institute of Health University Hospital, Bsalim, Lebanon
| | - Nazih Bizri
- Faculty of Medicine and Medical Sciences, 54686Universityof Balamand, Koura, Lebanon
| | - Nadine Francis
- Department of Internal Medicine, Middle East Institute of Health University Hospital, Bsalim, Lebanon
| | - Sabine Imad
- Department of Internal Medicine, Middle East Institute of Health University Hospital, Bsalim, Lebanon
| | - Maria Mezher
- Department of Internal Medicine, Middle East Institute of Health University Hospital, Bsalim, Lebanon
| | - Zahraa Mrad
- Department of Internal Medicine, Middle East Institute of Health University Hospital, Bsalim, Lebanon
| | - Jad Ibrahim
- Faculty of Sciences, 11238American University of Beirut, Beirut, Lebanon
| | - Kamal Zahran
- Hematology, Oncology and Bone Marrow Transplantation Unit, Middle East Institute of Health University Hospital, Bsalim, Lebanon
| | - Farah Farroukh
- Hematology, Oncology and Bone Marrow Transplantation Unit, Middle East Institute of Health University Hospital, Bsalim, Lebanon
| | - Malak Itani
- Hematology, Oncology and Bone Marrow Transplantation Unit, Middle East Institute of Health University Hospital, Bsalim, Lebanon
| | - Amani Assaad
- Faculty of Medicine, Lebanese University, Beirut, Lebanon
| | - Loubna Sinno
- Department of Medical Research, 36696Makassed General Hospital, Beirut, Lebanon
| | - Dania Abdallah
- Pharmacy Department, 36696Makassed General Hospital, Beirut, Lebanon
| | - Ahmad Ibrahim
- Faculty of Medicine, Lebanese University, Beirut, Lebanon.,Department of Internal Medicine, Division of Hematology, Oncology and Bone Marrow Transplantation, Middle East Institute of Health University Hospital, Bsalim, Lebanon.,Department of Internal Medicine, Division of Hematology, Oncology and Bone Marrow Transplantation, 36696Makassed General Hospital, Beirut, Lebanon
| |
Collapse
|
105
|
Rasalkar AA, Bhatia S, Katte T, Narayanan P, Vinjamuri S, Shettihalli AK, Kabade S, Manas RN, Kadappa V, Reddy DNS. COVID-19 and its impact on cancer, HIV, and mentally ill patients. LESSONS FROM COVID-19 2022. [PMCID: PMC9347297 DOI: 10.1016/b978-0-323-99878-9.00006-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2) and its disease, COVID-19 is a global pandemic creating an unprecedented medical as well economic havoc across the world. Despite the wide spread global infection rates, the death rate is low for COVID-19. However, COVID-19 patients with other comorbid conditions face severe health complications irrespective of their gender or age. As the management of COVID-19 patients is taking up health resources, it is getting difficult to treat patients suffering from other dreadful diseases like cancer, HIV, and mental health issues. In this chapter, we discuss the effects of COVID-19 and management of cancer patients of main cancer subtypes (e.g., breast, lung, blood cancers), and patients affected with HIV and mental health issues. Finally, we also add a perspective on Ayurvedic treatment and its efficacy on COVID-19 patients.
Collapse
|
106
|
Paudel S, Ghimire L, Jin L, Jeansonne D, Jeyaseelan S. Regulation of emergency granulopoiesis during infection. Front Immunol 2022; 13:961601. [PMID: 36148240 PMCID: PMC9485265 DOI: 10.3389/fimmu.2022.961601] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
During acute infectious and inflammatory conditions, a large number of neutrophils are in high demand as they are consumed in peripheral organs. The hematopoietic system rapidly responds to the demand by turning from steady state to emergency granulopoiesis to expedite neutrophil generation in the bone marrow (BM). How the hematopoietic system integrates pathogenic and inflammatory stress signals into the molecular cues of emergency granulopoiesis has been the subject of investigations. Recent studies in the field have highlighted emerging concepts, including the direct sensing of pathogens by BM resident or sentinel hematopoietic stem and progenitor cells (HSPCs), the crosstalk of HSPCs, endothelial cells, and stromal cells to convert signals to granulopoiesis, and the identification of novel inflammatory molecules, such as C/EBP-β, ROS, IL-27, IFN-γ, CXCL1 with direct effects on HSPCs. In this review, we will provide a detailed account of emerging concepts while reassessing well-established cellular and molecular players of emergency granulopoiesis. While providing our views on the discrepant results and theories, we will postulate an updated model of granulopoiesis in the context of health and disease.
Collapse
Affiliation(s)
- Sagar Paudel
- Center for Lung Biology and Disease, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Pathobiological Sciences, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Laxman Ghimire
- Center for Lung Biology and Disease, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Pathobiological Sciences, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Liliang Jin
- Center for Lung Biology and Disease, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Pathobiological Sciences, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Duane Jeansonne
- Center for Lung Biology and Disease, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Pathobiological Sciences, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Samithamby Jeyaseelan
- Center for Lung Biology and Disease, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Pathobiological Sciences, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States.,Section of Pulmonary and Critical Care, Department of Medicine, LSU Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
107
|
OUP accepted manuscript. Brief Funct Genomics 2022; 21:159-176. [DOI: 10.1093/bfgp/elac002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 11/14/2022] Open
|
108
|
Liu B, Tao C, Wu Z, Yao H, Wang DA. Engineering strategies to achieve efficient in vitro expansion of haematopoietic stem cells: development and improvement. J Mater Chem B 2022; 10:1734-1753. [DOI: 10.1039/d1tb02706a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Haematopoietic stem cells are the basis for building and maintaining lifelong haematopoietic mechanisms and important resources for the treatment of blood disorders. Haematopoietic niches are microenvironment in the body where...
Collapse
|
109
|
Papachristoforou E, Ramachandran P. Macrophages as key regulators of liver health and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 368:143-212. [PMID: 35636927 DOI: 10.1016/bs.ircmb.2022.04.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Macrophages are a heterogeneous population of innate immune cells and key cellular components of the liver. Hepatic macrophages consist of embryologically-derived resident Kupffer cells (KC), recruited monocyte-derived macrophages (MDM) and capsular macrophages. Both the diversity and plasticity of hepatic macrophage subsets explain their different functions in the maintenance of hepatic homeostasis and in injury processes in acute and chronic liver diseases. In this review, we assess the evidence for macrophage involvement in regulating both liver health and injury responses in liver diseases including acute liver injury (ALI), chronic liver disease (CLD) (including liver fibrosis) and hepatocellular carcinoma (HCC). In healthy livers, KC display critical functions such as phagocytosis, danger signal recognition, cytokine release, antigen processing and the ability to orchestrate immune responses and maintain immunological tolerance. However, in most liver diseases there is a striking hepatic MDM expansion, which orchestrate both disease progression and regression. Single-cell approaches have transformed our understanding of liver macrophage heterogeneity, dynamics, and functions in both human samples and preclinical models. We will further discuss the new insights provided by these approaches and how they are enabling high-fidelity work to specifically identify pathogenic macrophage subpopulations. Given the important role of macrophages in regulating injury responses in a broad range of settings, there is now a huge interest in developing new therapeutic strategies aimed at targeting macrophages. Therefore, we also review the current approaches being used to modulate macrophage function in liver diseases and discuss the therapeutic potential of targeting macrophage subpopulations as a novel treatment strategy for patients with liver disorders.
Collapse
Affiliation(s)
- Eleni Papachristoforou
- University of Edinburgh Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, Edinburgh, United Kingdom
| | - Prakash Ramachandran
- University of Edinburgh Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, Edinburgh, United Kingdom.
| |
Collapse
|
110
|
Zbinden A, Canté-Barrett K, Pike-Overzet K, Staal FJT. Stem Cell-Based Disease Models for Inborn Errors of Immunity. Cells 2021; 11:cells11010108. [PMID: 35011669 PMCID: PMC8750661 DOI: 10.3390/cells11010108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 11/24/2022] Open
Abstract
The intrinsic capacity of human hematopoietic stem cells (hHSCs) to reconstitute myeloid and lymphoid lineages combined with their self-renewal capacity hold enormous promises for gene therapy as a viable treatment option for a number of immune-mediated diseases, most prominently for inborn errors of immunity (IEI). The current development of such therapies relies on disease models, both in vitro and in vivo, which allow the study of human pathophysiology in great detail. Here, we discuss the current challenges with regards to developmental origin, heterogeneity and the subsequent implications for disease modeling. We review models based on induced pluripotent stem cell technology and those relaying on use of adult hHSCs. We critically review the advantages and limitations of current models for IEI both in vitro and in vivo. We conclude that existing and future stem cell-based models are necessary tools for developing next generation therapies for IEI.
Collapse
|
111
|
Haltalli MLR, Wilkinson AC, Rodriguez-Fraticelli A, Porteus M. Hematopoietic stem cell gene editing and expansion: State-of-the-art technologies and recent applications. Exp Hematol 2021; 107:9-13. [PMID: 34973360 DOI: 10.1016/j.exphem.2021.12.399] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/29/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) is a curative therapy for a range of hematological diseases, from leukemias to immunodeficiencies and anemias. The aim in using HSCT is to replace a patient's dysfunctional blood system with a functional one by transplanting healthy hematopoietic stem cells (HSCs). HSCs may be collected from a healthy donor (for allogeneic HSCT) or from the patient for genetic correction (for autologous HSCT gene therapies). Despite the curative potential of HSCT, several hurdles to its wider and safer use remain, including how to efficiently genetically correct HSCs and how to increase donor HSC numbers to improve the donor pool. In recent years, the development of state-of-the-art technologies, such as Cas9-AAV6 technologies and identification of the small molecule HSC agonist UM171, have accelerated progress in HSC gene editing and expansion. These translational research efforts were the focus of the Spring 2021 International Society for Experimental Hematology (ISEH) webinar. Here we present a summary and discussion of the implications of these new approaches to improve HSC-based therapy.
Collapse
Affiliation(s)
- Myriam L R Haltalli
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
| | - Adam C Wilkinson
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | - Matthew Porteus
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
112
|
Ren Y, Zhu J, Han Y, Li P, Wu J, Qu H, Zhang Z, Fang X. Regulatory association of long noncoding RNAs and chromatin accessibility facilitates erythroid differentiation. Blood Adv 2021; 5:5396-5409. [PMID: 34644394 PMCID: PMC9153002 DOI: 10.1182/bloodadvances.2021005167] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/13/2021] [Indexed: 01/17/2023] Open
Abstract
Erythroid differentiation is a dynamic process regulated by multiple factors, whereas the interaction between long noncoding RNAs (lncRNAs) and chromatin accessibility and its influence on erythroid differentiation remains unclear. To elucidate this interaction, we used hematopoietic stem cells, multipotent progenitor cells, common myeloid progenitor cells, megakaryocyte-erythroid progenitor cells, and erythroblasts from human cord blood as an erythroid differentiation model to explore the coordinated regulatory functions of lncRNAs and chromatin accessibility by integrating RNA-seq and ATAC-seq data. We revealed that the integrated network of chromatin accessibility and lncRNAs exhibits stage-specific changes throughout the erythroid differentiation process and that the changes at the erythroblast stage of maturation are dramatic. We identified a subset of stage-specific lncRNAs and transcription factors (TFs) that associate with chromatin accessibility during erythroid differentiation, in which lncRNAs are key regulators of terminal erythroid differentiation via an lncRNA-TF-gene network. LncRNA PCED1B-AS1 was revealed to regulate terminal erythroid differentiation by coordinating GATA1 dynamically binding to the chromatin and interacting with the cytoskeleton network during erythroid differentiation. DANCR, another lncRNA that is highly expressed at the megakaryocyte-erythroid progenitor cell stage, was verified to promote erythroid differentiation by compromising megakaryocyte differentiation and coordinating with chromatin accessibility and TFs, such as RUNX1. Overall, our results identify the associated network of lncRNAs and chromatin accessibility in erythropoiesis and provide novel insights into erythroid differentiation and abundant resources for further study.
Collapse
Affiliation(s)
| | - Junwei Zhu
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
| | - Yuanyuan Han
- Guizhou University, Medical College, Guiyang, China, China; and
| | - Pin Li
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
| | - Jing Wu
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| | - Hongzhu Qu
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
| | | | - Xiangdong Fang
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
113
|
Human Cerebral Organoid Implantation Alleviated the Neurological Deficits of Traumatic Brain Injury in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6338722. [PMID: 34853630 PMCID: PMC8629662 DOI: 10.1155/2021/6338722] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) causes a high rate of mortality and disability, and its treatment is still limited. Loss of neurons in damaged area is hardly rescued by relative molecular therapies. Based on its disease characteristics, we transplanted human embryonic stem cell- (hESC-) derived cerebral organoids in the brain lesions of controlled cortical impact- (CCI-) modeled severe combined immunodeficient (SCID) mice. Grafted organoids survived and differentiated in CCI-induced lesion pools in mouse cortical tissue. Implanted cerebral organoids differentiated into various types of neuronal cells, extended long projections, and showed spontaneous action, as indicated by electromyographic activity in the grafts. Induced vascularization and reduced glial scar were also found after organoid implantation, suggesting grafting could improve local situation and promote neural repair. More importantly, the CCI mice's spatial learning and memory improved after organoid grafting. These findings suggest that cerebral organoid implanted in lesion sites differentiates into cortical neurons, forms long projections, and reverses deficits in spatial learning and memory, a potential therapeutic avenue for TBI.
Collapse
|
114
|
Li J, Wang X, Ding J, Zhu Y, Min W, Kuang W, Yuan K, Sun C, Yang P. Development and clinical advancement of small molecules for ex vivo expansion of hematopoietic stem cell. Acta Pharm Sin B 2021; 12:2808-2831. [PMID: 35755294 PMCID: PMC9214065 DOI: 10.1016/j.apsb.2021.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/02/2021] [Accepted: 12/09/2021] [Indexed: 02/08/2023] Open
Abstract
Hematopoietic stem cell (HSC) transplantation is the only curative therapy for many diseases. HSCs from umbilical cord blood (UCB) source have many advantages over from bone marrow. However, limited HSC dose in a single CB unit restrict its widespread use. Over the past two decades, ex vivo HSC expansion with small molecules has been an effective approach for obtaining adequate HSCs. Till now, several small-molecule compounds have entered the phase I/II trials, showing safe and favorable pharmacological profiles. As HSC expansion has become a hot topic over recent years, many newly identified small molecules along with novel biological mechanisms for HSC expansion would help solve this challenging issue. Here, we will give an overview of HSC biology, discovery and medicinal chemistry development of small molecules, natural products targeting for HSC expansion, and their recent clinical progresses, as well as potential protein targets for HSC expansion.
Collapse
|
115
|
Malouf C, Loughran SJ, Wilkinson AC, Shimamura A, Río P. Translational research for bone marrow failure patients. Exp Hematol 2021; 105:18-21. [PMID: 34801643 DOI: 10.1016/j.exphem.2021.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/13/2021] [Indexed: 12/19/2022]
Abstract
Bone marrow failure syndromes encompass a range of inherited and acquired hematological diseases that result in insufficient blood cell production, which leads to severe complications including anemia, weakening of the immune system, impaired coagulation, and increased risk of cancer. Within inherited bone marrow failure syndromes, a number of genetically distinct diseases have been described including Shwachman-Diamond syndrome and Fanconi anemia. Given the genetic complexity and poor prognosis of these inherited bone marrow failure syndromes, there is increasing interest in both characterizing the genetic landscapes of these diseases and developing novel gene therapies to effectively monitor and cure patients. These topics were the focus of the winter 2021 International Society for Experimental Hematology New Investigator Webinar, which featured presentations by Dr. Akiko Shimamura and Dr. Paula Río. Here, we review the topics covered within this webinar.
Collapse
Affiliation(s)
- Camille Malouf
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Stephen J Loughran
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Adam C Wilkinson
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Akiko Shimamura
- Bone Marrow Failure and Myelodysplastic Syndrome Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Paula Río
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), Madrid, Spain; Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM), Madrid, Spain
| |
Collapse
|
116
|
Bone marrow microenvironment of MPN cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021. [PMID: 34756245 DOI: 10.1016/bs.ircmb.2021.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
In this chapter, we will discuss the current knowledge concerning the alterations of the cellular components in the bone marrow niche in Myeloproliferative Neoplasms (MPNs), highlighting the central role of the megakaryocytes in MPN progression, and the extracellular matrix components characterizing the fibrotic bone marrow.
Collapse
|
117
|
Rodriguez-Echeverri C, Bonilla-Porras A, Gonzalez A. Hematopoietic and Mesenchymal Stromal Cells: New Immunological Roles During Fungal Infections. Stem Cells Dev 2021; 30:1049-1055. [PMID: 34514808 DOI: 10.1089/scd.2021.0211] [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: 11/12/2022] Open
Abstract
Adult stem cells are characterized not only by their regenerative and immunomodulatory capacity but also by their therapeutic potential in various pathologies that include hematological malignancies, cancer, and autoimmune and inflammatory diseases, among others. However, these cells seem to play a paradoxical role during the development of the immune response in some infectious diseases. As an example, Candida albicans can induce the proliferation and differentiation of hematopoietic stem cells (HSCs) and their progenitors, a process known as emergency hematopoiesis. Moreover, Aspergillus fumigatus and C. albicans, once recognized by mesenchymal stromal cells (MSCs), can induce an anti-inflammatory or proinflammatory profile, respectively, and, in turn, these cells can inhibit the growth of these fungal pathogens. Additionally, the transplantation of MSCs, in an experimental pulmonary model of paracoccidioidomycosis, has been shown to exacerbate the inflammatory response. More recently, in vitro studies have shown that MSCs recognize Paracoccidioides brasiliensis through a mechanism mediated by toll-like receptor (TLR)2, TLR4, and Dectin-1, which, in turn, induces a proinflammatory profile. This review describes the main mechanisms and immunomodulatory properties of HSCs and MSCs during infections caused by some medically important fungal pathogens described so far in literature.
Collapse
Affiliation(s)
- Carolina Rodriguez-Echeverri
- Basic and Applied Microbiology Research Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellín, Colombia
| | - Angelica Bonilla-Porras
- Basic and Applied Microbiology Research Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellín, Colombia
| | - Angel Gonzalez
- Basic and Applied Microbiology Research Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
118
|
Challen GA, Pietras EM, Wallscheid NC, Signer RAJ. Simplified murine multipotent progenitor isolation scheme: Establishing a consensus approach for multipotent progenitor identification. Exp Hematol 2021; 104:55-63. [PMID: 34648848 DOI: 10.1016/j.exphem.2021.09.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 11/18/2022]
Abstract
The mouse hematopoietic system has served as a paradigm for analysis of developmental fate decisions in tissue homeostasis and regeneration. However, multiple immunophenotypic definitions of, and sometimes divergent nomenclatures used to classify, murine multipotent progenitors (MPPs) have emerged in the field over time. This has created significant confusion and inconsistency in the hematology field. To facilitate easier comparison of murine MPP phenotypes between research laboratories, a working group of four International Society for Experimental Hematology (ISEH) members with extensive experience studying the functional activities associated with different MPP phenotypic definitions reviewed the current state of the field with the goal of developing a position statement toward a simplified and unified immunophenotypic definition of MPP populations. In November of 2020, this position statement was presented as a webinar to the ISEH community for discussion and feedback. Hence, the Simplified MPP Identification Scheme presented here is the result of curation of existing literature, consultation with leaders in the field, and crowdsourcing from the wider experimental hematology community. Adoption of a unified definition and nomenclature, while still leaving room for individual investigator customization, will benefit scientists at all levels trying to compare these populations between experimental settings.
Collapse
Affiliation(s)
- Grant A Challen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Eric M Pietras
- Division of Hematology, Department of Medicine, Department of Microbiology and Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | - Robert A J Signer
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA
| |
Collapse
|
119
|
Acevedo OA, Berrios RV, Rodríguez-Guilarte L, Lillo-Dapremont B, Kalergis AM. Molecular and Cellular Mechanisms Modulating Trained Immunity by Various Cell Types in Response to Pathogen Encounter. Front Immunol 2021; 12:745332. [PMID: 34671359 PMCID: PMC8521023 DOI: 10.3389/fimmu.2021.745332] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/15/2021] [Indexed: 12/24/2022] Open
Abstract
The induction of trained immunity represents an emerging concept defined as the ability of innate immune cells to acquire a memory phenotype, which is a typical hallmark of the adaptive response. Key points modulated during the establishment of trained immunity include epigenetic, metabolic and functional changes in different innate-immune and non-immune cells. Regarding to epigenetic changes, it has been described that long non-coding RNAs (LncRNAs) act as molecular scaffolds to allow the assembly of chromatin-remodeling complexes that catalyze epigenetic changes on chromatin. On the other hand, relevant metabolic changes that occur during this process include increased glycolytic rate and the accumulation of metabolites from the tricarboxylic acid (TCA) cycle, which subsequently regulate the activity of histone-modifying enzymes that ultimately drive epigenetic changes. Functional consequences of established trained immunity include enhanced cytokine production, increased antigen presentation and augmented antimicrobial responses. In this article, we will discuss the current knowledge regarding the ability of different cell subsets to acquire a trained immune phenotype and the molecular mechanisms involved in triggering such a response. This knowledge will be helpful for the development of broad-spectrum therapies against infectious diseases based on the modulation of epigenetic and metabolic cues regulating the development of trained immunity.
Collapse
Affiliation(s)
- Orlando A. Acevedo
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roslye V. Berrios
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Linmar Rodríguez-Guilarte
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bastián Lillo-Dapremont
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
120
|
Flora P, Dalal G, Cohen I, Ezhkova E. Polycomb Repressive Complex(es) and Their Role in Adult Stem Cells. Genes (Basel) 2021; 12:1485. [PMID: 34680880 PMCID: PMC8535826 DOI: 10.3390/genes12101485] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/13/2021] [Accepted: 09/22/2021] [Indexed: 12/31/2022] Open
Abstract
Populations of resident stem cells (SCs) are responsible for maintaining, repairing, and regenerating adult tissues. In addition to having the capacity to generate all the differentiated cell types of the tissue, adult SCs undergo long periods of quiescence within the niche to maintain themselves. The process of SC renewal and differentiation is tightly regulated for proper tissue regeneration throughout an organisms' lifetime. Epigenetic regulators, such as the polycomb group (PcG) of proteins have been implicated in modulating gene expression in adult SCs to maintain homeostatic and regenerative balances in adult tissues. In this review, we summarize the recent findings that elucidate the composition and function of the polycomb repressive complex machinery and highlight their role in diverse adult stem cell compartments.
Collapse
Affiliation(s)
- Pooja Flora
- Department of Cell, Developmental, and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA;
| | - Gil Dalal
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel;
| | - Idan Cohen
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel;
| | - Elena Ezhkova
- Department of Cell, Developmental, and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA;
| |
Collapse
|
121
|
Bedel A, Boutin J, Amintas S, Lamrissi-Garcia I, Rousseau B, Poglio S, Brunet de la Grange P, Moranvillier I, Blouin JM, Richard E, Moreau-Gaudry F, Dabernat S. Spleen route accelerates engraftment of human hematopoietic stem cells. Biochem Biophys Res Commun 2021; 569:23-28. [PMID: 34216994 DOI: 10.1016/j.bbrc.2021.06.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 10/21/2022]
Abstract
Intravenous injections of human hematopoietic stem cells (hHSCs) is routinely used in clinic and for modeling hematopoiesis in mice. However, unspecific dilution in vascular system and non-hematopoietic organs challenges engraftment efficiency. Although spleen is capable of extra medullar hematopoiesis, its ability to support human HSC transplantation has never been evaluated. We demonstrate that intra-splenic injection results in high and sustained engraftment of hHSCs into immune-deficient mice, with higher chimerisms than with intravenous or intra-femoral injections. Our results support that spleen microenvironment provides a niche for HSCs amplification and offers a new route for efficient HSC transplantation.
Collapse
Affiliation(s)
- A Bedel
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France
| | - J Boutin
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France
| | - S Amintas
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France
| | - I Lamrissi-Garcia
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France
| | - B Rousseau
- Université de Bordeaux, Bordeaux, France; Animalerie spécialisée, Université de Bordeaux, Bordeaux, France
| | - S Poglio
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France
| | - P Brunet de la Grange
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; Etablissement Français du Sang-Aquitaine Limousin (EFS-AqLi), Bordeaux, France
| | - I Moranvillier
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France
| | - J M Blouin
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France
| | - E Richard
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France
| | - F Moreau-Gaudry
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France.
| | - S Dabernat
- Université de Bordeaux, Bordeaux, France; INSERM U1035, Bordeaux, France; CHU de Bordeaux, Bordeaux, France
| |
Collapse
|
122
|
Dight J, Zhao J, Styke C, Khosrotehrani K, Patel J. Resident vascular endothelial progenitor definition and function: the age of reckoning. Angiogenesis 2021; 25:15-33. [PMID: 34499264 PMCID: PMC8813834 DOI: 10.1007/s10456-021-09817-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/05/2021] [Indexed: 02/07/2023]
Abstract
The cardiovascular system is composed around the central function of the endothelium that lines the inner surfaces of its vessels. In recent years, the existence of a progenitor population within the endothelium has been validated through the study of endothelial colony-forming cells (ECFCs) in human peripheral blood and certain vascular beds. However, our knowledge on endothelial populations in vivo that can give rise to ECFCs in culture has been limited. In this review we report and analyse recent attempts at describing progenitor populations in vivo from murine studies that reflect the self-renewal and stemness capacity observed in ECFCs. We pinpoint seminal discoveries within the field, which have phenotypically defined, and functionally scrutinised these endothelial progenitors. Furthermore, we review recent publications utilising single-cell sequencing technologies to better understand the endothelium in homeostasis and pathology.
Collapse
Affiliation(s)
- James Dight
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - Jilai Zhao
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - Cassandra Styke
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - Kiarash Khosrotehrani
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia.
| | - Jatin Patel
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia. .,Cancer and Ageing Research Program, School of Biomedical Sciences, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia.
| |
Collapse
|
123
|
Wu F, Chen Z, Liu J, Hou Y. The Akt-mTOR network at the interface of hematopoietic stem cell homeostasis. Exp Hematol 2021; 103:15-23. [PMID: 34464661 DOI: 10.1016/j.exphem.2021.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/15/2022]
Abstract
Hematopoietic stem cells (HSCs) are immature blood cells that exhibit multilineage differentiation capacity. Homeostasis is critical for HSC potential and lifelong hematopoiesis, and HSC homeostasis is tightly governed by both intrinsic molecular networks and microenvironmental signals. The evolutionarily conserved serine/threonine protein kinase B (PKB, also referred to as Akt)-mammalian target of rapamycin (mTOR) pathway is universal to nearly all multicellular organisms and plays an integral role in most cellular processes. Emerging evidence has revealed a central role of the Akt-mTOR network in HSC homeostasis, because it responds to multiple intracellular and extracellular signals and regulates various downstream targets, eventually affecting several cellular processes, including the cell cycle, mitochondrial metabolism, and protein synthesis. Dysregulated Akt-mTOR signaling greatly affects HSC self-renewal, maintenance, differentiation, survival, autophagy, and aging, as well as transformation of HSCs to leukemia stem cells. Here, we review recent works and provide an advanced understanding of how the Akt-mTOR network regulates HSC homeostasis, thus offering insights into future clinical applications.
Collapse
Affiliation(s)
- Feng Wu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Zhe Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jingbo Liu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China.
| | - Yu Hou
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
124
|
Mannino G, Russo C, Maugeri G, Musumeci G, Vicario N, Tibullo D, Giuffrida R, Parenti R, Lo Furno D. Adult stem cell niches for tissue homeostasis. J Cell Physiol 2021; 237:239-257. [PMID: 34435361 PMCID: PMC9291197 DOI: 10.1002/jcp.30562] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/26/2021] [Accepted: 08/09/2021] [Indexed: 12/13/2022]
Abstract
Adult stem cells are fundamental to maintain tissue homeostasis, growth, and regeneration. They reside in specialized environments called niches. Following activating signals, they proliferate and differentiate into functional cells that are able to preserve tissue physiology, either to guarantee normal turnover or to counteract tissue damage caused by injury or disease. Multiple interactions occur within the niche between stem cell‐intrinsic factors, supporting cells, the extracellular matrix, and signaling pathways. Altogether, these interactions govern cell fate, preserving the stem cell pool, and regulating stem cell proliferation and differentiation. Based on their response to body needs, tissues can be largely classified into three main categories: tissues that even in normal conditions are characterized by an impressive turnover to replace rapidly exhausting cells (blood, epidermis, or intestinal epithelium); tissues that normally require only a basal cell replacement, though able to efficiently respond to increased tissue needs, injury, or disease (skeletal muscle); tissues that are equipped with less powerful stem cell niches, whose repairing ability is not able to overcome severe damage (heart or nervous tissue). The purpose of this review is to describe the main characteristics of stem cell niches in these different tissues, highlighting the various components influencing stem cell activity. Although much has been done, more work is needed to further increase our knowledge of niche interactions. This would be important not only to shed light on this fundamental chapter of human physiology but also to help the development of cell‐based strategies for clinical therapeutic applications, especially when other approaches fail.
Collapse
Affiliation(s)
- Giuliana Mannino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Cristina Russo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
125
|
Yao CL, Huang P, Liu TC, Lin YK, Chen CY, Lai YT, Chin TY, Tseng TY, Hsu YC. CCL2 associated with CD38 expression during ex vivo expansion in human cord blood-derived hematopoietic stem cells. Aging (Albany NY) 2021; 13:19878-19893. [PMID: 34375303 PMCID: PMC8386547 DOI: 10.18632/aging.203398] [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] [Received: 02/22/2021] [Accepted: 07/08/2021] [Indexed: 11/26/2022]
Abstract
To date, different experimental strategies have been developed for the ex vivo expansion of human hematopoietic stem cells (HSCs) for clinical applications. However, differences in the genomic function of expanded HSCs under different culture systems remain unclear. In this study, we compared the gene expression profiles of HSCs in ex vivo expanded serum (10% FBS, fetal bovine serum) and serum-free culture systems and analyzed the molecular functions of differentially expressed genes using microarray chips. We identified 839 differentially expressed genes between the two culture systems. These genes were enriched in the TNF -regulated inflammatory pathway in an FBS culture system. In addition, the mRNA expression of CCL2 (C-C motif chemokine ligand 2), TNF (tumor necrosis factor) and FOS (FBJ murine osteosarcoma viral oncogene homolog) was validated by RT-qPCR. Our data revealed that ex vivo expansion of HSCs using the FBS culture system induces an inflammatory response and high CD38 expression, indicating that this system might activate an inflammatory pathway and induce expression of the cancer marker CD38 during ex vivo expansion of HSCs. This study provides a transcriptional profile and new insights into the genomic functions of HSCs under different expanded cultures.
Collapse
Affiliation(s)
- Chao-Ling Yao
- Department of Chemical Engineering, National Cheng Kung University, Tainan 701, Taiwan.,Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li, Taoyuan City 320, Taiwan.,Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Chung-Li, Taoyuan City 320, Taiwan
| | - Poyin Huang
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City 807, Taiwan.,Department of Neurology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung City 807, Taiwan.,Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung City 807, Taiwan.,Department of Neurology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
| | - Tsai-Chi Liu
- Department of Biomedical Sciences and Engineering, National Central University, Chung-Li, Taoyuan City 320, Taiwan
| | - Yung-Kai Lin
- Institute of Food Safety and Risk Management, National Taiwan Ocean University, Keelung City 202, Taiwan.,Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung City 402, Taiwan
| | - Ching-Yun Chen
- Department of Biomedical Sciences and Engineering, National Central University, Chung-Li, Taoyuan City 320, Taiwan
| | - Yi-Ting Lai
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li, Taoyuan City 320, Taiwan
| | - Tzu Yun Chin
- Department of Biomedical Sciences and Engineering, National Central University, Chung-Li, Taoyuan City 320, Taiwan
| | - Tsung-Yu Tseng
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Chung-Li, Taoyuan City 320, Taiwan
| | - Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Chung-Li, Taoyuan City 320, Taiwan
| |
Collapse
|
126
|
ANP32B-mediated repression of p53 contributes to maintenance of normal and CML stem cells. Blood 2021; 138:2485-2498. [PMID: 34359074 DOI: 10.1182/blood.2020010400] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 08/03/2021] [Indexed: 11/20/2022] Open
Abstract
Proper regulation of p53 signaling is critical for the maintenance of hematopoietic stem cells (HSCs) and leukemic stem cells (LSCs). The hematopoietic cell-specific mechanisms regulating p53 activity remain largely unknown. Here, we demonstrate that conditional deletion of acidic leucine-rich nuclear phosphoprotein 32B (ANP32B) in hematopoietic cells impairs repopulation capacity and post-injury regeneration of HSCs. Mechanistically, ANP32B forms a repressive complex with and thus inhibits the transcriptional activity of p53 in hematopoietic cells, and p53 deletion rescues the functional defect in Anp32b-deficient HSCs. Of great interest, ANP32B is highly expressed in leukemic cells from chronic myelogenous leukemia (CML) patients. Anp32b deletion enhances p53 transcriptional activity to impair LSCs function in a murine CML model, and exhibits synergistic therapeutic effects with tyrosine kinase inhibitors in inhibiting CML propagation. In summary, our findings provide a novel strategy to enhance p53 activity in LSCs by inhibiting ANP32B, and identify ANP32B as a potential therapeutic target in treating CML.
Collapse
|
127
|
Therapeutic targeting of TGF-β in cancer: hacking a master switch of immune suppression. Clin Sci (Lond) 2021; 135:35-52. [PMID: 33399850 PMCID: PMC7796313 DOI: 10.1042/cs20201236] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/26/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
Abstract
Cancers may escape elimination by the host immune system by rewiring the tumour microenvironment towards an immune suppressive state. Transforming growth factor-β (TGF-β) is a secreted multifunctional cytokine that strongly regulates the activity of immune cells while, in parallel, can promote malignant features such as cancer cell invasion and migration, angiogenesis, and the emergence of cancer-associated fibroblasts. TGF-β is abundantly expressed in cancers and, most often, its abundance associated with poor clinical outcomes. Immunotherapeutic strategies, particularly T cell checkpoint blockade therapies, so far, only produce clinical benefit in a minority of cancer patients. The inhibition of TGF-β activity is a promising approach to increase the efficacy of T cell checkpoint blockade therapies. In this review, we briefly outline the immunoregulatory functions of TGF-β in physiological and malignant contexts. We then deliberate on how the therapeutic targeting of TGF-β may lead to a broadened applicability and success of state-of-the-art immunotherapies.
Collapse
|
128
|
Udroiu I, Sgura A. Growing and aging of hematopoietic stem cells. World J Stem Cells 2021; 13:594-604. [PMID: 34249229 PMCID: PMC8246248 DOI: 10.4252/wjsc.v13.i6.594] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/22/2020] [Accepted: 02/02/2021] [Indexed: 02/06/2023] Open
Abstract
In the hematopoietic system, a small number of stem cells produce a progeny of several distinct lineages. During ontogeny, they arise in the aorta-gonad-mesonephros region of the embryo and the placenta, afterwards colonise the liver and finally the bone marrow. After this fetal phase of rapid expansion, the number of hematopoietic stem cells continues to grow, in order to sustain the increasing blood volume of the developing newborn, and eventually reaches a steady-state. The kinetics of this growth are mirrored by the rates of telomere shortening in leukocytes. During adulthood, hematopoietic stem cells undergo a very small number of cell divisions. Nonetheless, they are subjected to aging, eventually reducing their potential to produce differentiated progeny. The causal relationships between telomere shortening, DNA damage, epigenetic changes, and aging have still to be elucidated.
Collapse
Affiliation(s)
- Ion Udroiu
- Department of Science, Roma Tre University, Rome 00146, Italy
| | - Antonella Sgura
- Department of Science, Roma Tre University, Rome 00146, Italy
| |
Collapse
|
129
|
Oedekoven CA, Belmonte M, Bode D, Hamey FK, Shepherd MS, Che JLC, Boyd G, McDonald C, Belluschi S, Diamanti E, Bastos HP, Bridge KS, Göttgens B, Laurenti E, Kent DG. Hematopoietic stem cells retain functional potential and molecular identity in hibernation cultures. Stem Cell Reports 2021; 16:1614-1628. [PMID: 33961793 PMCID: PMC8190576 DOI: 10.1016/j.stemcr.2021.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 02/02/2023] Open
Abstract
Advances in the isolation and gene expression profiling of single hematopoietic stem cells (HSCs) have permitted in-depth resolution of their molecular program. However, long-term HSCs can only be isolated to near purity from adult mouse bone marrow, thereby precluding studies of their molecular program in different physiological states. Here, we describe a powerful 7-day HSC hibernation culture system that maintains HSCs as single cells in the absence of a physical niche. Single hibernating HSCs retain full functional potential compared with freshly isolated HSCs with respect to colony-forming capacity and transplantation into primary and secondary recipients. Comparison of hibernating HSC molecular profiles to their freshly isolated counterparts showed a striking degree of molecular similarity, further resolving the core molecular machinery of HSC self-renewal while also identifying key factors that are potentially dispensable for HSC function, including members of the AP1 complex (Jun, Fos, and Ncor2), Sult1a1 and Cish. Finally, we provide evidence that hibernating mouse HSCs can be transduced without compromising their self-renewal activity and demonstrate the applicability of hibernation cultures to human HSCs.
Collapse
Affiliation(s)
- Caroline A Oedekoven
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK
| | - Miriam Belmonte
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK
| | - Daniel Bode
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK
| | - Fiona K Hamey
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK
| | - Mairi S Shepherd
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK
| | - James Lok Chi Che
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK
| | - Grace Boyd
- York Biomedical Research Institute, Department of Biology, University of York, York YO10 5DD, UK
| | - Craig McDonald
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK
| | - Serena Belluschi
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK
| | - Evangelia Diamanti
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK
| | - Hugo P Bastos
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK
| | - Katherine S Bridge
- York Biomedical Research Institute, Department of Biology, University of York, York YO10 5DD, UK
| | - Berthold Göttgens
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK
| | - Elisa Laurenti
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK
| | - David G Kent
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK; York Biomedical Research Institute, Department of Biology, University of York, York YO10 5DD, UK.
| |
Collapse
|
130
|
Baghbani E, Noorolyai S, Duijf PHG, Silvestris N, Kolahian S, Hashemzadeh S, Baghbanzadeh Kojabad A, FallahVazirabad A, Baradaran B. The impact of microRNAs on myeloid-derived suppressor cells in cancer. Hum Immunol 2021; 82:668-678. [PMID: 34020831 DOI: 10.1016/j.humimm.2021.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 02/08/2023]
Abstract
Inflammation promotes cancer development. To a large extent, this can be attributed to the recruitment of myeloid-derived suppressor cells (MDSCs) to tumors. These cells are known for establishing an immunosuppressive tumor microenvironment by suppressing T cell activities. However, MDSCs also promote metastasis and angiogenesis. Critically, as small non-coding RNAs that regulate gene expression, microRNAs (miRNAs) control MDSC activities. In this review, we discuss how miRNA networks regulate key MDSC signaling pathways, how they shape MDSC development, differentiation and activation, and how this impacts tumor development. By targeting the expression of miRNAs in MDSCs, we can alter their main signaling pathways. In turn, this can compromise their ability to promote multiple hallmarks of cancer. Therefore, this may represent a new powerful strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Noorolyai
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal H G Duijf
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Australia; University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Nicola Silvestris
- IRCCS Bari, Italy. Medical Oncology Unit-IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy, Department of Biomedical Sciences and Human Oncology DIMO-University of Bari, Bari, Italy
| | - Saeed Kolahian
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Division of Pharmacogenomics, University of Tübingen, Tübingen, Germany; Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University of Marburg, Marburg, Germany; Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Marburg, Germany
| | - Shahryar Hashemzadeh
- General and Vascular Surgery Department, Imam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
131
|
Sommerkamp P, Cabezas-Wallscheid N, Trumpp A. Alternative Polyadenylation in Stem Cell Self-Renewal and Differentiation. Trends Mol Med 2021; 27:660-672. [PMID: 33985920 DOI: 10.1016/j.molmed.2021.04.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022]
Abstract
Cellular function is shaped by transcriptional and post-transcriptional mechanisms, including alternative polyadenylation (APA). By directly controlling 3'- untranslated region (UTR) length and the selection of the last exon, APA regulates up to 70% of all cellular transcripts influencing RNA stability, output, and protein isoform expression. Cell-state-dependent 3'-UTR shortening has been identified as a hallmark of cellular proliferation. Hence, quiescent/dormant stem cells are characterized by long 3'-UTRs, whereas proliferative stem/progenitor cells exhibit 3'-UTR shortening. Here, the latest studies analyzing the role of APA in regulating stem cell state, self-renewal, differentiation, and metabolism are reviewed. The new role of APA in controlling stem cell fate opens novel potential therapeutic avenues in the field of regenerative medicine.
Collapse
Affiliation(s)
- Pia Sommerkamp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | | | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69117 Heidelberg, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany.
| |
Collapse
|
132
|
Dong Y, Yang Q, Sun X. Comprehensive Analysis of Cell Therapy on Chronic Skin Wound Healing: A Meta-Analysis. Hum Gene Ther 2021; 32:787-795. [PMID: 33446038 DOI: 10.1089/hum.2020.275] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Wound healing has been greatly challenging in different acute and chronic skin injuries. Among them, nonrevascularizable critical limb ischemic ulcers, venous leg ulcers, and diabetic lower limb or extremity ulcers are well-known refractory skin injuries that are difficult to treat. Partly differentiated, progenitor cell-based graft transplantation or direct injection of autologous stem cells might promote the wound healing process. Studies aiming to comprehensively analyze the effects of cell therapy on skin wound healing could provide clinical evidence for skin injury treatment. Different databases were searched for full-text publications on the comparison between cell therapy and regular therapy. Heterogeneity was detected by the I2 method, and a fixed effect model was applied for data pooling if heterogeneity was absent. Publication bias was analyzed using a funnel plot, and 10 studies were finally included in this study. After a long-term follow-up, fewer patients underwent major amputation in the cell therapy group, compared with the standard therapy group, and those in the cell therapy group were characterized by a smaller ulcer area. Moreover, there was a significant difference in the wound healing rate between the intervention and control groups. However, pain caused by skin wounds was hardly mitigated by cell therapy in patients with critical limb ischemia. In this study, cell therapy proved effective in decreasing the size of ulcers and improving the wound closure rate. Additionally, the major amputation rate was decreased in the cell therapy group. However, the symptoms of pain were hardly alleviated by cell therapy in patients with cutaneous ulcers caused by peripheral artery disease-related critical limb ischemia.
Collapse
Affiliation(s)
- Yujie Dong
- Departments of Emergency, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qi Yang
- Departments of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaojie Sun
- Plastic and Reconstruction Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
133
|
McKenna DH, Stroncek DF. Cellular Engineering. Transfus Med 2021. [DOI: 10.1002/9781119599586.ch19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
134
|
Chen Z, Huo D, Li L, Liu Z, Li Z, Xu S, Huang Y, Wu W, Zhou C, Liu Y, Kuang M, Wu F, Li H, Qian P, Song G, Wu X, Chen J, Hou Y. Nuclear DEK preserves hematopoietic stem cells potential via NCoR1/HDAC3-Akt1/2-mTOR axis. J Exp Med 2021; 218:e20201974. [PMID: 33755722 PMCID: PMC7992411 DOI: 10.1084/jem.20201974] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/16/2021] [Accepted: 02/18/2021] [Indexed: 12/14/2022] Open
Abstract
The oncogene DEK is found fused with the NUP214 gene creating oncoprotein DEK-NUP214 that induces acute myeloid leukemia (AML) in patients, and secreted DEK protein functions as a hematopoietic cytokine to regulate hematopoiesis; however, the intrinsic role of nuclear DEK in hematopoietic stem cells (HSCs) remains largely unknown. Here, we show that HSCs lacking DEK display defects in long-term self-renew capacity, eventually resulting in impaired hematopoiesis. DEK deficiency reduces quiescence and accelerates mitochondrial metabolism in HSCs, in part, dependent upon activating mTOR signaling. At the molecular level, DEK recruits the corepressor NCoR1 to repress acetylation of histone 3 at lysine 27 (H3K27ac) and restricts the chromatin accessibility of HSCs, governing the expression of quiescence-associated genes (e.g., Akt1/2, Ccnb2, and p21). Inhibition of mTOR activity largely restores the maintenance and potential of Dek-cKO HSCs. These findings highlight the crucial role of nuclear DEK in preserving HSC potential, uncovering a new link between chromatin remodelers and HSC homeostasis, and have clinical implications.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dawei Huo
- Department of Cell Biology, Tianjin Medical University, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Tianjin, China
| | - Lei Li
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhilong Liu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhigang Li
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shuangnian Xu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yongxiu Huang
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Weiru Wu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chengfang Zhou
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuanyuan Liu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Mei Kuang
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Feng Wu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hui Li
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Pengxu Qian
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Xudong Wu
- Department of Cell Biology, Tianjin Medical University, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Tianjin, China
| | - Jieping Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yu Hou
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
135
|
Raz AA, Wurtzel O, Reddien PW. Planarian stem cells specify fate yet retain potency during the cell cycle. Cell Stem Cell 2021; 28:1307-1322.e5. [PMID: 33882291 DOI: 10.1016/j.stem.2021.03.021] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 01/08/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
Planarian whole-body regeneration is enabled by stem cells called neoblasts. At least some neoblasts are individually pluripotent. Neoblasts are also heterogeneous, with subpopulations of specialized neoblasts having different specified fates. Fate specification in neoblasts is regulated by fate-specific transcription factor (FSTF) expression. Here, we find that FSTF expression is common in neoblast S/G2/M cell-cycle phases but less common in G1. We find that specialized neoblasts can divide to produce progeny with asymmetric cell fates, suggesting that they could retain pluripotency. Furthermore, no known neoblast class was present in all neoblast colonies, suggesting that pluripotency is not the exclusive property of any known class. We tested this possibility with single-cell transplantations, which indicate that at least some specialized neoblasts are likely clonogenic. On the basis of these findings, we propose a model for neoblast pluripotency in which neoblasts can undergo specialization during the cell cycle without loss of potency.
Collapse
Affiliation(s)
- Amelie A Raz
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Omri Wurtzel
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Peter W Reddien
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
136
|
Multistage feedback-driven compartmental dynamics of hematopoiesis. iScience 2021; 24:102326. [PMID: 33889822 PMCID: PMC8050392 DOI: 10.1016/j.isci.2021.102326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 02/02/2021] [Accepted: 03/15/2021] [Indexed: 11/22/2022] Open
Abstract
Human hematopoiesis is surprisingly resilient to disruptions, providing suitable responses to severe bleeding, long-lasting immune activation, and even bone marrow transplants. Still, many blood disorders exist which push the system past its natural plasticity, resulting in abnormalities in the circulating blood. While proper treatment of such diseases can benefit from understanding the underlying cell dynamics, these are non-trivial to predict due to the hematopoietic system's hierarchical nature and complex feedback networks. To characterize the dynamics following different types of perturbations, we investigate a model representing hematopoiesis as a sequence of compartments covering all maturation stages-from stem to mature cells-where feedback regulates cell production to ongoing necessities. We find that a stable response to perturbations requires the simultaneous adaptation of cell differentiation and self-renewal rates, and show that under conditions of continuous disruption-as found in chronic hemolytic states-compartment cell numbers evolve to novel stable states.
Collapse
|
137
|
Ijabi J, Afrisham R, Moradi-Sardareh H, Roozehdar P, Seifi F, Sahebkar A, Ijabi R. The Shift of HbF to HbA under Influence of SKA2 Gene; A Possible Link between Cortisol and Hematopoietic Maturation in Term and Preterm Newborns. Endocr Metab Immune Disord Drug Targets 2021; 21:485-494. [PMID: 32364083 DOI: 10.2174/1871530320666200504091354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND We hypothesized that the SKA2 gene can convert hemoglobin F to A leading to the maturity of the hematopoietic system by glucocorticoid hormone; so, the present study aimed to investigate the health outcome of newborns by using the effect of SKA2 gene on hematopoietic maturation. METHODS At first, 142 samples were divided into term and preterm. After sampling from the umbilical cord blood, the expression of SKA2 genes and HbA and F were evaluated by quantitative RT-PCR. The blood gases were measured by Campact 3 device. Finally, the cortisol level was measured by ELISA method and HbA and F levels were investigated by capillary electrophoresis. RESULTS The blood gases and Apgar scores were more favorable in term newborns (P <0.001). Levels of protein/expression of HbF in newborns with Apgar score greater than 7 was lower than that of the newborns with Apgar score below 7 (P <0.001). Cortisol and HbA levels were considerably higher in term newborns compared to the preterm ones (P <0.001). In the preterm and term groups, SKA2 gene expression had a positive and significant relationship with cortisol and HbA levels as well as a negative relationship with the HbF level. In the preterm group, a positive and significant relationship was observed between the expression of SKA2 and HbF genes. CONCLUSION The results revealed that the SKA2 gene affected hematopoietic maturation in preterm and term newborns and the health outcome of newborns improved by increasing HbA level.
Collapse
Affiliation(s)
- Janat Ijabi
- Department of Hematology, School of Allied Health, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Afrisham
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hemen Moradi-Sardareh
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Roozehdar
- Department of Medical Veterinary, Azad University, Garmsar Branch, Garmsar, Iran
| | - Fatemeh Seifi
- Counseling and Reproductive Health Research Centre, Golestan University of Medical Sciences, Gorgan, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Roghayeh Ijabi
- Counseling and Reproductive Health Research Centre, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
138
|
Lana JF, da Fonseca LF, Azzini G, Santos G, Braga M, Cardoso Junior AM, Murrell WD, Gobbi A, Purita J, Percope de Andrade MA. Bone Marrow Aspirate Matrix: A Convenient Ally in Regenerative Medicine. Int J Mol Sci 2021; 22:ijms22052762. [PMID: 33803231 PMCID: PMC7963152 DOI: 10.3390/ijms22052762] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/17/2021] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
The rise in musculoskeletal disorders has prompted medical experts to devise novel effective alternatives to treat complicated orthopedic conditions. The ever-expanding field of regenerative medicine has allowed researchers to appreciate the therapeutic value of bone marrow-derived biological products, such as the bone marrow aspirate (BMA) clot, a potent orthobiologic which has often been dismissed and regarded as a technical complication. Numerous in vitro and in vivo studies have contributed to the expansion of medical knowledge, revealing optimistic results concerning the application of autologous bone marrow towards various impactful disorders. The bone marrow accommodates a diverse family of cell populations and a rich secretome; therefore, autologous BMA-derived products such as the “BMA Matrix”, may represent a safe and viable approach, able to reduce the costs and some drawbacks linked to the expansion of bone marrow. BMA provides —it eliminates many hurdles associated with its preparation, especially in regards to regulatory compliance. The BMA Matrix represents a suitable alternative, indicated for the enhancement of tissue repair mechanisms by modulating inflammation and acting as a natural biological scaffold as well as a reservoir of cytokines and growth factors that support cell activity. Although promising, more clinical studies are warranted in order to further clarify the efficacy of this strategy.
Collapse
Affiliation(s)
- José Fábio Lana
- IOC—Instituto do Osso e da Cartilagem, 1386 Presidente Kennedy Avenue, Indaiatuba 13334-170, Brazil; (J.F.L.); (G.A.)
| | | | - Gabriel Azzini
- IOC—Instituto do Osso e da Cartilagem, 1386 Presidente Kennedy Avenue, Indaiatuba 13334-170, Brazil; (J.F.L.); (G.A.)
| | - Gabriel Santos
- IOC—Instituto do Osso e da Cartilagem, 1386 Presidente Kennedy Avenue, Indaiatuba 13334-170, Brazil; (J.F.L.); (G.A.)
- Correspondence:
| | - Marcelo Braga
- Hospital São Judas Tadeu, 150 Cel. João Notini St, Divinópolis 35500-017, Brazil;
| | - Alvaro Motta Cardoso Junior
- Núcleo Avançado de Estudos em Ortopedia e Neurocirurgia, 2144 Ibirapuera Avenue, São Paulo 04028-001, Brazil;
| | - William D. Murrell
- Abu Dhabi Knee and Sports Medicine, Healthpoint Hospital, Zayed Sports City, Between Gate 1 and 6, Abu Dhabi 00000 (P. O. Box No. 112308), United Arab Emirates;
- 411th Hospital Center, Bldg 938, Birmingham Ave, Naval Air Station, Jacksonville, FL 32212, USA
| | - Alberto Gobbi
- O.A.S.I. Bioresearch Foundation Gobbi Onlus, 20133 Milano, Italy;
| | - Joseph Purita
- Institute of Regenerative Medicine, Boca Raton, FL 33432, USA;
| | | |
Collapse
|
139
|
Chabi S, To THV, Leavitt R, Poglio S, Jorge PG, Jaccard M, Petersson K, Petit B, Roméo PH, Pflumio F, Vozenin MC, Uzan B. Ultra-high-dose-rate FLASH and Conventional-Dose-Rate Irradiation Differentially Affect Human Acute Lymphoblastic Leukemia and Normal Hematopoiesis. Int J Radiat Oncol Biol Phys 2021; 109:819-829. [PMID: 33075474 DOI: 10.1016/j.ijrobp.2020.10.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/30/2020] [Accepted: 10/09/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE Ultra-high-dose-rate FLASH radiation therapy has been shown to minimize side effects of irradiation in various organs while keeping antitumor efficacy. This property, called the FLASH effect, has caused enthusiasm in the radiation oncology community because it opens opportunities for safe dose escalation and improved radiation therapy outcome. Here, we investigated the impact of ultra-high-dose-rate FLASH versus conventional-dose-rate (CONV) total body irradiation (TBI) on humanized models of T-cell acute lymphoblastic leukemia (T-ALL) and normal human hematopoiesis. METHODS AND MATERIALS We optimized the geometry of irradiation to ensure reproducible and homogeneous procedures using eRT6/Oriatron. Three T-ALL patient-derived xenografts and hematopoietic stem/progenitor cells (HSPCs) and CD34+ cells isolated from umbilical cord blood were transplanted into immunocompromised mice, together or separately. After reconstitution, mice received 4 Gy FLASH and CONV-TBI, and tumor growth and normal hematopoiesis were studied. A retrospective study of clinical and gene-profiling data previously obtained on the 3 T-ALL patient-derived xenografts was performed. RESULTS FLASH-TBI was more efficient than CONV-TBI in controlling the propagation of 2 cases of T-ALL, whereas the third case of T-ALL was more responsive to CONV-TBI. The 2 FLASH-sensitive cases of T-ALL had similar genetic abnormalities, and a putative susceptibility imprint to FLASH-RT was found. In addition, FLASH-TBI was able to preserve some HSPC/CD34+ cell potential. Interestingly, when HSPC and T-ALL were present in the same animals, FLASH-TBI could control tumor development in most (3 of 4) of the secondary grafted animals, whereas among the mice receiving CONV-TBI, treated cells died with high leukemia infiltration. CONCLUSIONS Compared with CONV-TBI, FLASH-TBI reduced functional damage to human blood stem cells and had a therapeutic effect on human T-ALL with a common genetic and genomic profile. The validity of the defined susceptibility imprint needs to be investigated further; however, to our knowledge, the present findings are the first to show benefits of FLASH-TBI on human hematopoiesis and leukemia treatment.
Collapse
Affiliation(s)
- Sara Chabi
- Team Niche and Cancer in Hematopoiesis, Fontenay-aux-Roses, France; Laboratoire des cellules Souches Hématopoïétiques et des Leucémies, Service Cellules Souches et Radiations, Fontenay-aux-Roses, France; UMRE008 Stabilité Génétique, Cellules Souches et Radiations, Université de Paris and Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Thi Hong Van To
- Laboratoire des cellules Souches Hématopoïétiques et des Leucémies, Service Cellules Souches et Radiations, Fontenay-aux-Roses, France; UMRE008 Stabilité Génétique, Cellules Souches et Radiations, Université de Paris and Université Paris-Saclay, Fontenay-aux-Roses, France; Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Ron Leavitt
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Sandrine Poglio
- Team Niche and Cancer in Hematopoiesis, Fontenay-aux-Roses, France; Laboratoire des cellules Souches Hématopoïétiques et des Leucémies, Service Cellules Souches et Radiations, Fontenay-aux-Roses, France; UMRE008 Stabilité Génétique, Cellules Souches et Radiations, Université de Paris and Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Patrik Gonçalves Jorge
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland; Institute of Radiation Physics/CHUV, Lausanne University Hospital, Switzerland
| | - Maud Jaccard
- Institute of Radiation Physics/CHUV, Lausanne University Hospital, Switzerland
| | - Kristoffer Petersson
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland; Institute of Radiation Physics/CHUV, Lausanne University Hospital, Switzerland
| | - Benoit Petit
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Paul-Henri Roméo
- Team Niche and Cancer in Hematopoiesis, Fontenay-aux-Roses, France; UMRE008 Stabilité Génétique, Cellules Souches et Radiations, Université de Paris and Université Paris-Saclay, Fontenay-aux-Roses, France; Laboratoire de la Régulation de la Transcription dans les cellules Souches, Service Cellules Souches et Radiations, Fontenay-aux-Roses, France
| | - Françoise Pflumio
- Team Niche and Cancer in Hematopoiesis, Fontenay-aux-Roses, France; Laboratoire des cellules Souches Hématopoïétiques et des Leucémies, Service Cellules Souches et Radiations, Fontenay-aux-Roses, France; UMRE008 Stabilité Génétique, Cellules Souches et Radiations, Université de Paris and Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Marie-Catherine Vozenin
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland.
| | - Benjamin Uzan
- Team Niche and Cancer in Hematopoiesis, Fontenay-aux-Roses, France; Laboratoire des cellules Souches Hématopoïétiques et des Leucémies, Service Cellules Souches et Radiations, Fontenay-aux-Roses, France; UMRE008 Stabilité Génétique, Cellules Souches et Radiations, Université de Paris and Université Paris-Saclay, Fontenay-aux-Roses, France
| |
Collapse
|
140
|
Horton PD, Dumbali SP, Bhanu KR, Diaz MF, Wenzel PL. Biomechanical Regulation of Hematopoietic Stem Cells in the Developing Embryo. CURRENT TISSUE MICROENVIRONMENT REPORTS 2021; 2:1-15. [PMID: 33937868 PMCID: PMC8087251 DOI: 10.1007/s43152-020-00027-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 12/16/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The contribution of biomechanical forces to hematopoietic stem cell (HSC) development in the embryo is a relatively nascent area of research. Herein, we address the biomechanics of the endothelial-to-hematopoietic transition (EHT), impact of force on organelles, and signaling triggered by extrinsic forces within the aorta-gonad-mesonephros (AGM), the primary site of HSC emergence. RECENT FINDINGS Hemogenic endothelial cells undergo carefully orchestrated morphological adaptations during EHT. Moreover, expansion of the stem cell pool during embryogenesis requires HSC extravasation into the circulatory system and transit to the fetal liver, which is regulated by forces generated by blood flow. Findings from other cell types also suggest that forces external to the cell are sensed by the nucleus and mitochondria. Interactions between these organelles and the actin cytoskeleton dictate processes such as cell polarization, extrusion, division, survival, and differentiation. SUMMARY Despite challenges of measuring and modeling biophysical cues in the embryonic HSC niche, the past decade has revealed critical roles for mechanotransduction in governing HSC fate decisions. Lessons learned from the study of the embryonic hematopoietic niche promise to provide critical insights that could be leveraged for improvement in HSC generation and expansion ex vivo.
Collapse
Affiliation(s)
- Paulina D. Horton
- Department of Integrative Biology & Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 4.130, Houston, TX 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Immunology Program, MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Sandeep P. Dumbali
- Department of Integrative Biology & Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 4.130, Houston, TX 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Krithikaa Rajkumar Bhanu
- Immunology Program, MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Miguel F. Diaz
- Department of Integrative Biology & Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 4.130, Houston, TX 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Pamela L. Wenzel
- Department of Integrative Biology & Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 4.130, Houston, TX 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Immunology Program, MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
141
|
Guijarro-Hernández A, Vizmanos JL. A Broad Overview of Signaling in Ph-Negative Classic Myeloproliferative Neoplasms. Cancers (Basel) 2021; 13:cancers13050984. [PMID: 33652860 PMCID: PMC7956519 DOI: 10.3390/cancers13050984] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary There is growing evidence that Ph-negative myeloproliferative neoplasms are disorders in which multiple signaling pathways are significantly disturbed. The heterogeneous phenotypes observed among patients have highlighted the importance of having a comprehensive knowledge of the molecular mechanisms behind these diseases. This review aims to show a broad overview of the signaling involved in myeloproliferative neoplasms (MPNs) and other processes that can modify them, which could be helpful to better understand these diseases and develop more effective targeted treatments. Abstract Ph-negative myeloproliferative neoplasms (polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF)) are infrequent blood cancers characterized by signaling aberrations. Shortly after the discovery of the somatic mutations in JAK2, MPL, and CALR that cause these diseases, researchers extensively studied the aberrant functions of their mutant products. In all three cases, the main pathogenic mechanism appears to be the constitutive activation of JAK2/STAT signaling and JAK2-related pathways (MAPK/ERK, PI3K/AKT). However, some other non-canonical aberrant mechanisms derived from mutant JAK2 and CALR have also been described. Moreover, additional somatic mutations have been identified in other genes that affect epigenetic regulation, tumor suppression, transcription regulation, splicing and other signaling pathways, leading to the modification of some disease features and adding a layer of complexity to their molecular pathogenesis. All of these factors have highlighted the wide variety of cellular processes and pathways involved in the pathogenesis of MPNs. This review presents an overview of the complex signaling behind these diseases which could explain, at least in part, their phenotypic heterogeneity.
Collapse
Affiliation(s)
- Ana Guijarro-Hernández
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain;
| | - José Luis Vizmanos
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain;
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Correspondence:
| |
Collapse
|
142
|
Belmonte RL, Engbretson IL, Kim JH, Cajias I, Ahn EYE, Stachura DL. son is necessary for proper vertebrate blood development. PLoS One 2021; 16:e0247489. [PMID: 33630943 PMCID: PMC7906411 DOI: 10.1371/journal.pone.0247489] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
The gene SON is on human chromosome 21 (21q22.11) and is thought to be associated with hematopoietic disorders that accompany Down syndrome. Additionally, SON is an RNA splicing factor that plays a role in the transcription of leukemia-associated genes. Previously, we showed that mutations in SON cause malformations in human and zebrafish spines and brains during early embryonic development. To examine the role of SON in normal hematopoiesis, we reduced expression of the zebrafish homolog of SON in zebrafish at the single-cell developmental stage with specific morpholinos. In addition to the brain and spinal malformations we also observed abnormal blood cell levels upon son knockdown. We then investigated how blood production was altered when levels of son were reduced. Decreased levels of son resulted in lower amounts of red blood cells when visualized with lcr:GFP transgenic fish. There were also reduced thrombocytes seen with cd41:GFP fish, and myeloid cells when mpx:GFP fish were examined. We also observed a significant decrease in the quantity of T cells, visualized with lck:GFP fish. However, when we examined their hematopoietic stem and progenitor cells (HSPCs), we saw no difference in colony-forming capability. These studies indicate that son is essential for the proper differentiation of the innate and adaptive immune system, and further investigation determining the molecular pathways involved during blood development should elucidate important information about vertebrate HSPC generation, proliferation, and differentiation.
Collapse
Affiliation(s)
- Rebecca L. Belmonte
- Department of Biological Sciences, California State University Chico, Chico, California, United States of America
| | - Isabella L. Engbretson
- Department of Biological Sciences, California State University Chico, Chico, California, United States of America
| | - Jung-Hyun Kim
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, United States of America
| | - Illiana Cajias
- Department of Biological Sciences, California State University Chico, Chico, California, United States of America
| | - Eun-Young Erin Ahn
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - David L. Stachura
- Department of Biological Sciences, California State University Chico, Chico, California, United States of America
- * E-mail:
| |
Collapse
|
143
|
Sonoda Y. Human CD34-negative hematopoietic stem cells: The current understanding of their biological nature. Exp Hematol 2021; 96:13-26. [PMID: 33610645 DOI: 10.1016/j.exphem.2021.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 01/29/2021] [Accepted: 02/07/2021] [Indexed: 12/29/2022]
Abstract
Hematopoietic stem cell (HSC) heterogeneity and hierarchy are a current topic of interest, having major implications for clinical HSC transplantation and basic research on human HSCs. It was long believed that the most primitive HSCs in mammals, including mice and humans, were CD34 antigen positive (CD34+). However, 2 decades ago, it was reported that murine long-term multilineage reconstituting HSCs were lineage marker negative (Lin-, i.e., c-kit+Sca-1+CD34low/-), known as CD34low/- KSL cells. In contrast, human CD34- HSCs, a counterpart of murine CD34low/- KSL cells, were hard to identify for a long time mainly because of their rarity. We previously identified very primitive human cord blood (CB)-derived CD34- severe combined immunodeficiency (SCID)-repopulating cells (SRCs) using the intra-bone marrow injection method and proposed the new concept that CD34- SRCs (HSCs) reside at the apex of the human HSC hierarchy. Through a series of studies, we identified two positive/enrichment markers: CD133 and GPI-80. The combination of these two markers enabled the development of an ultrahigh-resolution purification method for CD34- as well as CD34+ HSCs and the successful purification of both HSCs at the single-cell level. Cell population purity is a crucial prerequisite for reliable biological and molecular analyses. Clonal analyses of highly purified human CD34- HSCs have revealed their potent megakaryocyte/erythrocyte differentiation potential. Based on these observations, we propose a revised road map for the commitment of human CB-derived CD34- HSCs. This review updates the current understanding of the stem cell nature of human CB-derived primitive CD34- as well as CD34+ HSCs.
Collapse
Affiliation(s)
- Yoshiaki Sonoda
- Department of iPS Stem Cell Regenerative Medicine, Kansai Medical University, Osaka, Japan.
| |
Collapse
|
144
|
Jackson TR, Ling RE, Roy A. The Origin of B-cells: Human Fetal B Cell Development and Implications for the Pathogenesis of Childhood Acute Lymphoblastic Leukemia. Front Immunol 2021; 12:637975. [PMID: 33679795 PMCID: PMC7928347 DOI: 10.3389/fimmu.2021.637975] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/28/2021] [Indexed: 12/27/2022] Open
Abstract
Human B-lymphopoiesis is a dynamic life-long process that starts in utero by around six post-conception weeks. A detailed understanding of human fetal B-lymphopoiesis and how it changes in postnatal life is vital for building a complete picture of normal B-lymphoid development through ontogeny, and its relevance in disease. B-cell acute lymphoblastic leukemia (B-ALL) is one of the most common cancers in children, with many of the leukemia-initiating events originating in utero. It is likely that the biology of B-ALL, including leukemia initiation, maintenance and progression depends on the developmental stage and type of B-lymphoid cell in which it originates. This is particularly important for early life leukemias, where specific characteristics of fetal B-cells might be key to determining how the disease behaves, including response to treatment. These cellular, molecular and/or epigenetic features are likely to change with age in a cell intrinsic and/or microenvironment directed manner. Most of our understanding of fetal B-lymphopoiesis has been based on murine data, but many recent studies have focussed on characterizing human fetal B-cell development, including functional and molecular assays at a single cell level. In this mini-review we will give a short overview of the recent advances in the understanding of human fetal B-lymphopoiesis, including its relevance to infant/childhood leukemia, and highlight future questions in the field.
Collapse
Affiliation(s)
- Thomas R Jackson
- Department of Paediatrics and MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Rebecca E Ling
- Department of Paediatrics and MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Anindita Roy
- Department of Paediatrics and MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, United Kingdom
| |
Collapse
|
145
|
Höpner SS, Raykova A, Radpour R, Amrein MA, Koller D, Baerlocher GM, Riether C, Ochsenbein AF. LIGHT/LTβR signaling regulates self-renewal and differentiation of hematopoietic and leukemia stem cells. Nat Commun 2021; 12:1065. [PMID: 33594067 PMCID: PMC7887212 DOI: 10.1038/s41467-021-21317-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/17/2021] [Indexed: 12/30/2022] Open
Abstract
The production of blood cells during steady-state and increased demand depends on the regulation of hematopoietic stem cell (HSC) self-renewal and differentiation. Similarly, the balance between self-renewal and differentiation of leukemia stem cells (LSCs) is crucial in the pathogenesis of leukemia. Here, we document that the TNF receptor superfamily member lymphotoxin-β receptor (LTβR) and its ligand LIGHT regulate quiescence and self-renewal of murine and human HSCs and LSCs. Cell-autonomous LIGHT/LTβR signaling on HSCs reduces cell cycling, promotes symmetric cell division and prevents primitive HSCs from exhaustion in serial re-transplantation experiments and genotoxic stress. LTβR deficiency reduces the numbers of LSCs and prolongs survival in a murine chronic myeloid leukemia (CML) model. Similarly, LIGHT/LTβR signaling in human G-CSF mobilized HSCs and human LSCs results in increased colony forming capacity in vitro. Thus, our results define LIGHT/LTβR signaling as an important pathway in the regulation of the self-renewal of HSCs and LSCs.
Collapse
MESH Headings
- Animals
- Antigens, CD34/metabolism
- Cell Cycle/drug effects
- Cell Cycle/genetics
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cell Self Renewal/drug effects
- Cell Self Renewal/genetics
- DNA Damage
- Fluorouracil/pharmacology
- Gene Expression Regulation, Leukemic/drug effects
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Lymphotoxin beta Receptor/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction/drug effects
- Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
- Mice
Collapse
Affiliation(s)
- S S Höpner
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Ana Raykova
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - R Radpour
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - M A Amrein
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - D Koller
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - G M Baerlocher
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - C Riether
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - A F Ochsenbein
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
146
|
Morcos MNF, Zerjatke T, Glauche I, Munz CM, Ge Y, Petzold A, Reinhardt S, Dahl A, Anstee NS, Bogeska R, Milsom MD, Säwén P, Wan H, Bryder D, Roers A, Gerbaulet A. Continuous mitotic activity of primitive hematopoietic stem cells in adult mice. J Exp Med 2021; 217:151684. [PMID: 32302400 PMCID: PMC7971128 DOI: 10.1084/jem.20191284] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/23/2019] [Accepted: 03/04/2020] [Indexed: 01/04/2023] Open
Abstract
The proliferative activity of aging hematopoietic stem cells (HSCs) is controversially discussed. Inducible fluorescent histone 2B fusion protein (H2B-FP) transgenic mice are important tools for tracking the mitotic history of murine HSCs in label dilution experiments. A recent study proposed that primitive HSCs symmetrically divide only four times to then enter permanent quiescence. We observed that background fluorescence due to leaky H2B-FP expression, occurring in all H2B-FP transgenes independent of label induction, accumulated with age in HSCs with high repopulation potential. We argue that this background had been misinterpreted as stable retention of induced label. We found cell division–independent half-lives of H2B-FPs to be short, which had led to overestimation of HSC divisional activity. Our data do not support abrupt entry of HSCs into permanent quiescence or sudden loss of regeneration potential after four divisions, but show that primitive HSCs of adult mice continue to cycle rarely.
Collapse
Affiliation(s)
- Mina N F Morcos
- Institute for Immunology, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Thomas Zerjatke
- Institute for Medical Informatics and Biometry, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Ingmar Glauche
- Institute for Medical Informatics and Biometry, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Clara M Munz
- Institute for Immunology, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Yan Ge
- Institute for Immunology, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Andreas Petzold
- DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, TU Dresden, Dresden, Germany
| | - Susanne Reinhardt
- DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, TU Dresden, Dresden, Germany
| | - Andreas Dahl
- DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, TU Dresden, Dresden, Germany
| | - Natasha S Anstee
- Division of Experimental Hematology, Deutsches Krebsforschungszentrum and Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Heidelberg, Germany
| | - Ruzhica Bogeska
- Division of Experimental Hematology, Deutsches Krebsforschungszentrum and Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Heidelberg, Germany
| | - Michael D Milsom
- Division of Experimental Hematology, Deutsches Krebsforschungszentrum and Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Heidelberg, Germany
| | - Petter Säwén
- Division of Molecular Hematology, Lund University, Lund, Sweden
| | - Haixia Wan
- Division of Molecular Hematology, Lund University, Lund, Sweden
| | - David Bryder
- Division of Molecular Hematology, Lund University, Lund, Sweden.,Sahlgrenska Cancer Centre, Gothenburg University, Gothenburg, Sweden
| | - Axel Roers
- Institute for Immunology, Faculty of Medicine, TU Dresden, Dresden, Germany
| | | |
Collapse
|
147
|
Implications of hematopoietic stem cells heterogeneity for gene therapies. Gene Ther 2021; 28:528-541. [PMID: 33589780 PMCID: PMC8455331 DOI: 10.1038/s41434-021-00229-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 12/30/2020] [Accepted: 01/18/2021] [Indexed: 12/29/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) is the therapeutic concept to cure the blood/immune system of patients suffering from malignancies, immunodeficiencies, red blood cell disorders, and inherited bone marrow failure syndromes. Yet, allogeneic HSCT bear considerable risks for the patient such as non-engraftment, or graft-versus host disease. Transplanting gene modified autologous HSCs is a promising approach not only for inherited blood/immune cell diseases, but also for the acquired immunodeficiency syndrome. However, there is emerging evidence for substantial heterogeneity of HSCs in situ as well as ex vivo that is also observed after HSCT. Thus, HSC gene modification concepts are suggested to consider that different blood disorders affect specific hematopoietic cell types. We will discuss the relevance of HSC heterogeneity for the development and manufacture of gene therapies and in exemplary diseases with a specific emphasis on the key target HSC types myeloid-biased, lymphoid-biased, and balanced HSCs.
Collapse
|
148
|
Sommerkamp P, Mercier FE, Wilkinson AC, Bonnet D, Bourgine PE. Engineering human hematopoietic environments through ossicle and bioreactor technologies exploitation. Exp Hematol 2021; 94:20-25. [PMID: 33278488 PMCID: PMC7879567 DOI: 10.1016/j.exphem.2020.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/23/2020] [Accepted: 11/29/2020] [Indexed: 01/16/2023]
Abstract
The bone marrow microenvironment contains cellular niches that maintain the pool of hematopoietic stem and progenitor cells and support hematopoietic maturation. Malignant hematopoietic cells also co-opt normal cellular interactions to promote their own growth and evade therapy. In vivo systems used to study human hematopoiesis have been developed through transplantation into immunodeficient mouse models. However, incomplete cross-compatibility between the murine stroma and transplanted human hematopoietic cells limits the rate of engraftment and the study of relevant interactions. To supplement in vivo xenotransplantation models, complementary strategies have recently been developed, including the use of three-dimensional human bone marrow organoids in vivo, generated from bone marrow stromal cells seeded onto osteo-inductive scaffolds, as well as the use of ex vivo bioreactor models. These topics were the focus of the Spring 2020 International Society for Experimental Hematology New Investigator webinar. We review here the latest advances in generating humanized hematopoietic organoids and how they allow for the study of novel microenvironmental interactions.
Collapse
Affiliation(s)
- Pia Sommerkamp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - François E Mercier
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Adam C Wilkinson
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | - Dominique Bonnet
- The Francis Crick Institute, Haematopoietic Stem Cell Laboratory, London, UK
| | - Paul E Bourgine
- Laboratory for Cell, Tissue, and Organ Engineering, Department of Clinical Sciences, Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
149
|
Carreras P, González I, Gallardo M, Ortiz-Ruiz A, Morales ML, Encinas J, Martínez-López J. Long-Term Human Hematopoietic Stem Cell Culture in Microdroplets. MICROMACHINES 2021; 12:90. [PMID: 33467039 PMCID: PMC7830102 DOI: 10.3390/mi12010090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/16/2020] [Accepted: 01/12/2021] [Indexed: 12/28/2022]
Abstract
We previously reported a new approach for micromanipulation and encapsulation of human stem cells using a droplet-based microfluidic device. This approach demonstrated the possibility of encapsulating and culturing difficult-to-preserve primary human hematopoietic stem cells using an engineered double-layered bead composed by an inner layer of alginate and an outer layer of Puramatrix. We also demonstrated the maintenance and expansion of Multiple Myeloma cells in this construction. Here, the presented microfluidic technique is applied to construct a 3D biomimetic model to recapitulate the human hematopoietic stem cell niche using double-layered hydrogel beads cultured in 10% FBS culture medium. In this model, the long-term maintenance of the number of cells and expansion of hHSCS encapsulated in the proposed structures was observed. Additionally, a phenotypic characterization of the human hematopoietic stem cells generated in the presented biomimetic model was performed in order to assess their long-term stemness maintenance. Results indicate that the ex vivo cultured human CD34+ cells from bone marrow were viable, maintained, and expanded over a time span of eight weeks. This novel long-term stem cell culture methodology could represent a novel breakthrough to improve Hematopoietic Progenitor cell Transplant (HPT) as well as a novel tool for further study of the biochemical and biophysical factors influencing stem cell behavior. This technology opens a myriad of new applications as a universal stem cell niche model potentially able to expand other types of cells.
Collapse
Affiliation(s)
- Pilar Carreras
- CSIC, Spanish National Research Council, 28006 Madrid, Spain;
- Hospital 12 Octubre, Hematology Department, Research Institute i+12, 28040 Madrid, Spain; (M.G.); (A.O.-R.); (M.L.M.); (J.E.); (J.M.-L.)
| | - Itziar González
- CSIC, Spanish National Research Council, 28006 Madrid, Spain;
| | - Miguel Gallardo
- Hospital 12 Octubre, Hematology Department, Research Institute i+12, 28040 Madrid, Spain; (M.G.); (A.O.-R.); (M.L.M.); (J.E.); (J.M.-L.)
- CNIO, Spanish National Cancer Research Centre, Hematological Malignancies Research Unit, 28029 Madrid, Spain
| | - Alejandra Ortiz-Ruiz
- Hospital 12 Octubre, Hematology Department, Research Institute i+12, 28040 Madrid, Spain; (M.G.); (A.O.-R.); (M.L.M.); (J.E.); (J.M.-L.)
- CNIO, Spanish National Cancer Research Centre, Hematological Malignancies Research Unit, 28029 Madrid, Spain
| | - Maria Luz Morales
- Hospital 12 Octubre, Hematology Department, Research Institute i+12, 28040 Madrid, Spain; (M.G.); (A.O.-R.); (M.L.M.); (J.E.); (J.M.-L.)
- CNIO, Spanish National Cancer Research Centre, Hematological Malignancies Research Unit, 28029 Madrid, Spain
| | - Jessica Encinas
- Hospital 12 Octubre, Hematology Department, Research Institute i+12, 28040 Madrid, Spain; (M.G.); (A.O.-R.); (M.L.M.); (J.E.); (J.M.-L.)
- CNIO, Spanish National Cancer Research Centre, Hematological Malignancies Research Unit, 28029 Madrid, Spain
| | - Joaquín Martínez-López
- Hospital 12 Octubre, Hematology Department, Research Institute i+12, 28040 Madrid, Spain; (M.G.); (A.O.-R.); (M.L.M.); (J.E.); (J.M.-L.)
- CNIO, Spanish National Cancer Research Centre, Hematological Malignancies Research Unit, 28029 Madrid, Spain
- UCM, Medical Faculty, Complutense University Madrid, 28040 Madrid, Spain
| |
Collapse
|
150
|
Generation and manipulation of human iPSC-derived platelets. Cell Mol Life Sci 2021; 78:3385-3401. [PMID: 33439272 PMCID: PMC7804213 DOI: 10.1007/s00018-020-03749-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/01/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022]
Abstract
The discovery of iPSCs has led to the ex vivo production of differentiated cells for regenerative medicine. In the case of transfusion products, the derivation of platelets from iPSCs is expected to complement our current blood-donor supplied transfusion system through donor-independent production with complete pathogen-free assurance. This derivation can also overcome alloimmune platelet transfusion refractoriness by resulting in autologous, HLA-homologous or HLA-deficient products. Several developments were necessary to produce a massive number of platelets required for a single transfusion. First, expandable megakaryocytes were established from iPSCs through transgene expression. Second, a turbulent-type bioreactor with improved platelet yield and quality was developed. Third, novel drugs that enabled efficient feeder cell-free conditions were developed. Fourth, the platelet-containing suspension was purified and resuspended in an appropriate buffer. Finally, the platelet product needed to be assured for competency and safety including non-tumorigenicity through in vitro and in vivo preclinical tests. Based on these advancements, a clinical trial has started. The generation of human iPSC-derived platelets could evolve transfusion medicine to the next stage and assure a ubiquitous, safe supply of platelet products. Further, considering the feasibility of gene manipulations in iPSCs, other platelet products may bring forth novel therapeutic measures.
Collapse
|