1
|
Pereira AL, Galli S, Nombela‐Arrieta C. Bone marrow niches for hematopoietic stem cells. Hemasphere 2024; 8:e133. [PMID: 39086665 PMCID: PMC11289431 DOI: 10.1002/hem3.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/05/2024] [Accepted: 05/06/2024] [Indexed: 08/02/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are the cornerstone of the hematopoietic system. HSCs sustain the continuous generation of mature blood derivatives while self-renewing to preserve a relatively constant pool of progenitors throughout life. Yet, long-term maintenance of functional HSCs exclusively takes place in association with their native tissue microenvironment of the bone marrow (BM). HSCs have been long proposed to reside in fixed and identifiable anatomical units found in the complex BM tissue landscape, which control their identity and fate in a deterministic manner. In the last decades, tremendous progress has been made in the dissection of the cellular and molecular fabric of the BM, the structural organization governing tissue function, and the plethora of interactions established by HSCs. Nonetheless, a holistic model of the mechanisms controlling HSC regulation in their niche is lacking to date. Here, we provide an overview of our current understanding of BM anatomy, HSC localization, and crosstalk within local cellular neighborhoods in murine and human tissues, and highlight fundamental open questions on how HSCs functionally integrate in the BM microenvironment.
Collapse
Affiliation(s)
- Ana Luísa Pereira
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| | - Serena Galli
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| | - César Nombela‐Arrieta
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| |
Collapse
|
2
|
Hofmann J, Kokkaliaris KD. Bone marrow niches for hematopoietic stem cells: life span dynamics and adaptation to acute stress. Blood 2024; 144:21-34. [PMID: 38579285 DOI: 10.1182/blood.2023023788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/07/2024] Open
Abstract
ABSTRACT Hematopoietic stem cells (HSCs) are instrumental for organismal survival because they are responsible for lifelong production of mature blood lineages in homeostasis and response to external stress. To fulfill their function, HSCs rely on reciprocal interactions with specialized tissue microenvironments, termed HSC niches. From embryonic development to advanced aging, HSCs transition through several hematopoietic organs in which they are supported by distinct extrinsic cues. Here, we describe recent discoveries on how HSC niches collectively adapt to ensure robust hematopoietic function during biological aging and after exposure to acute stress. We also discuss the latest strategies leveraging niche-derived signals to revert aging-associated phenotypes and enhance hematopoietic recovery after myeloablation.
Collapse
Affiliation(s)
- Johanna Hofmann
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
- Department 15, Biosciences, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Konstantinos D Kokkaliaris
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Quantitative Spatial Cancer Biology Laboratory, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
- University Cancer Center, Frankfurt am Main, Germany
| |
Collapse
|
3
|
Tong Z, Yin Z. Distribution, contribution and regulation of nestin + cells. J Adv Res 2024; 61:47-63. [PMID: 37648021 PMCID: PMC11258671 DOI: 10.1016/j.jare.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Nestin is an intermediate filament first reported in neuroepithelial stem cells. Nestin expression could be found in a variety of tissues throughout all systems of the body, especially during tissue development and tissue regeneration processes. AIM OF REVIEW This review aimed to summarize and discuss current studies on the distribution, contribution and regulation of nestin+ cells in different systems of the body, to discuss the feasibility ofusing nestin as a marker of multilineage stem/progenitor cells, and better understand the potential roles of nestin+ cells in tissue development, regeneration and pathological processes. KEY SCIENTIFIC CONCEPTS OF REVIEW This review highlights the potential of nestin as a marker of multilineage stem/progenitor cells, and as a key factor in tissue development and tissue regeneration. The article discussed the current findings, limitations, and potential clinical implications or applications of nestin+ cells. Additionally, it included the relationship of nestin+ cells to other cell populations. We propose potential future research directions to encourage further investigation in the field.
Collapse
Affiliation(s)
- Ziyang Tong
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zi Yin
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| |
Collapse
|
4
|
Mendes M, Monteiro AC, Neto E, Barrias CC, Sobrinho-Simões MA, Duarte D, Caires HR. Transforming the Niche: The Emerging Role of Extracellular Vesicles in Acute Myeloid Leukaemia Progression. Int J Mol Sci 2024; 25:4430. [PMID: 38674015 PMCID: PMC11050723 DOI: 10.3390/ijms25084430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Acute myeloid leukaemia (AML) management remains a significant challenge in oncology due to its low survival rates and high post-treatment relapse rates, mainly attributed to treatment-resistant leukaemic stem cells (LSCs) residing in bone marrow (BM) niches. This review offers an in-depth analysis of AML progression, highlighting the pivotal role of extracellular vesicles (EVs) in the dynamic remodelling of BM niche intercellular communication. We explore recent advancements elucidating the mechanisms through which EVs facilitate complex crosstalk, effectively promoting AML hallmarks and drug resistance. Adopting a temporal view, we chart the evolving landscape of EV-mediated interactions within the AML niche, underscoring the transformative potential of these insights for therapeutic intervention. Furthermore, the review discusses the emerging understanding of endothelial cell subsets' impact across BM niches in shaping AML disease progression, adding another layer of complexity to the disease progression and treatment resistance. We highlight the potential of cutting-edge methodologies, such as organ-on-chip (OoC) and single-EV analysis technologies, to provide unprecedented insights into AML-niche interactions in a human setting. Leveraging accumulated insights into AML EV signalling to reconfigure BM niches and pioneer novel approaches to decipher the EV signalling networks that fuel AML within the human context could revolutionise the development of niche-targeted therapy for leukaemia eradication.
Collapse
Affiliation(s)
- Manuel Mendes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.M.); (A.C.M.); (E.N.); (C.C.B.); (M.A.S.-S.); (D.D.)
- ICBAS—Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Ana C. Monteiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.M.); (A.C.M.); (E.N.); (C.C.B.); (M.A.S.-S.); (D.D.)
- ICBAS—Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Estrela Neto
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.M.); (A.C.M.); (E.N.); (C.C.B.); (M.A.S.-S.); (D.D.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Cristina C. Barrias
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.M.); (A.C.M.); (E.N.); (C.C.B.); (M.A.S.-S.); (D.D.)
- ICBAS—Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Manuel A. Sobrinho-Simões
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.M.); (A.C.M.); (E.N.); (C.C.B.); (M.A.S.-S.); (D.D.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, 4200-135 Porto, Portugal
- Department of Clinical Haematology, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal
- Clinical Haematology, Department of Medicine, Faculdade de Medicina da Universidade do Porto (FMUP), 4200-319 Porto, Portugal
| | - Delfim Duarte
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.M.); (A.C.M.); (E.N.); (C.C.B.); (M.A.S.-S.); (D.D.)
- Unit of Biochemistry, Department of Biomedicine, Faculdade de Medicina da Universidade do Porto (FMUP), 4200-319 Porto, Portugal
- Department of Hematology and Bone Marrow Transplantation, Instituto Português de Oncologia (IPO)-Porto, 4200-072 Porto, Portugal
| | - Hugo R. Caires
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.M.); (A.C.M.); (E.N.); (C.C.B.); (M.A.S.-S.); (D.D.)
| |
Collapse
|
5
|
Sarachakov A, Varlamova A, Svekolkin V, Polyakova M, Valencia I, Unkenholz C, Pannellini T, Galkin I, Ovcharov P, Tabakov D, Postovalova E, Shin N, Sethi I, Bagaev A, Itkin T, Crane G, Kluk M, Geyer J, Inghirami G, Patel S. Spatial mapping of human hematopoiesis at single-cell resolution reveals aging-associated topographic remodeling. Blood 2023; 142:2282-2295. [PMID: 37774374 DOI: 10.1182/blood.2023021280] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/01/2023] Open
Abstract
ABSTRACT The spatial anatomy of hematopoiesis in the bone marrow (BM) has been extensively studied in mice and other preclinical models, but technical challenges have precluded a commensurate exploration in humans. Institutional pathology archives contain thousands of paraffinized BM core biopsy tissue specimens, providing a rich resource for studying the intact human BM topography in a variety of physiologic states. Thus, we developed an end-to-end pipeline involving multiparameter whole tissue staining, in situ imaging at single-cell resolution, and artificial intelligence-based digital whole slide image analysis and then applied it to a cohort of disease-free samples to survey alterations in the hematopoietic topography associated with aging. Our data indicate heterogeneity in marrow adipose tissue (MAT) content within each age group and an inverse correlation between MAT content and proportions of early myeloid and erythroid precursors, irrespective of age. We identify consistent endosteal and perivascular positioning of hematopoietic stem and progenitor cells (HSPCs) with medullary localization of more differentiated elements and, importantly, uncover new evidence of aging-associated changes in cellular and vascular morphologies, microarchitectural alterations suggestive of foci with increased lymphocytes, and diminution of a potentially active megakaryocytic niche. Overall, our findings suggest that there is topographic remodeling of human hematopoiesis associated with aging. More generally, we demonstrate the potential to deeply unravel the spatial biology of normal and pathologic human BM states using intact archival tissue specimens.
Collapse
Affiliation(s)
| | | | | | | | - Itzel Valencia
- Multiparametric In Situ Imaging Laboratory, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Caitlin Unkenholz
- Multiparametric In Situ Imaging Laboratory, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Tania Pannellini
- Multiparametric In Situ Imaging Laboratory, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | | | | | | | | | | | | | | | - Tomer Itkin
- Division of Regenerative Medicine, Department of Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY
| | - Genevieve Crane
- Department of Laboratory Medicine, Cleveland Clinic, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland, OH
| | - Michael Kluk
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine/NewYork-Presbyterian Hospital, New York, NY
| | - Julia Geyer
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine/NewYork-Presbyterian Hospital, New York, NY
| | - Giorgio Inghirami
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine/NewYork-Presbyterian Hospital, New York, NY
| | - Sanjay Patel
- Multiparametric In Situ Imaging Laboratory, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine/NewYork-Presbyterian Hospital, New York, NY
| |
Collapse
|
6
|
Takeishi S, Marchand T, Koba WR, Borger DK, Xu C, Guha C, Bergman A, Frenette PS, Gritsman K, Steidl U. Haematopoietic stem cell numbers are not solely determined by niche availability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.28.564559. [PMID: 37961493 PMCID: PMC10634881 DOI: 10.1101/2023.10.28.564559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Haematopoietic stem cells (HSCs) reside in specialized microenvironments, also referred to as niches, and it has been widely believed that HSC numbers are determined by the niche size alone 1-5 . However, the vast excess of the number of niche cells over that of HSCs raises questions about this model. We initially established a mathematical model of niche availability and occupancy, which predicted that HSC numbers are restricted at both systemic and local levels. To address this question experimentally, we developed a femoral bone transplantation system, enabling us to increase the number of available HSC niches. We found that the addition of niches does not alter total HSC numbers in the body, regardless of whether the endogenous (host) niche is intact or defective, suggesting that HSC numbers are limited at the systemic level. Additionally, HSC numbers in transplanted wild-type femurs did not increase beyond physiological levels when HSCs were mobilized from defective endogenous niches to the periphery, indicating that HSC numbers are also constrained at the local level. Our study demonstrates that HSC numbers are not solely determined by niche availability, thereby rewriting the long-standing model for the regulation of HSC numbers.
Collapse
|
7
|
Giallongo C, Dulcamare I, Giallongo S, Duminuco A, Pieragostino D, Cufaro MC, Amorini AM, Lazzarino G, Romano A, Parrinello N, Di Rosa M, Broggi G, Caltabiano R, Caraglia M, Scrima M, Pasquale LS, Tathode MS, Li Volti G, Motterlini R, Di Raimondo F, Tibullo D, Palumbo GA. MacroH2A1.1 as a crossroad between epigenetics, inflammation and metabolism of mesenchymal stromal cells in myelodysplastic syndromes. Cell Death Dis 2023; 14:686. [PMID: 37852977 PMCID: PMC10584900 DOI: 10.1038/s41419-023-06197-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 09/15/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023]
Abstract
Ineffective hematopoiesis is a hallmark of myelodysplastic syndromes (MDS). Hematopoietic alterations in MDS patients strictly correlate with microenvironment dysfunctions, eventually affecting also the mesenchymal stromal cell (MSC) compartment. Stromal cells are indeed epigenetically reprogrammed to cooperate with leukemic cells and propagate the disease as "tumor unit"; therefore, changes in MSC epigenetic profile might contribute to the hematopoietic perturbations typical of MDS. Here, we unveil that the histone variant macroH2A1 (mH2A1) regulates the crosstalk between epigenetics and inflammation in MDS-MSCs, potentially affecting their hematopoietic support ability. We show that the mH2A1 splicing isoform mH2A1.1 accumulates in MDS-MSCs, correlating with the expression of the Toll-like receptor 4 (TLR4), an important pro-tumor activator of MSC phenotype associated to a pro-inflammatory behavior. MH2A1.1-TLR4 axis was further investigated in HS-5 stromal cells after ectopic mH2A1.1 overexpression (mH2A1.1-OE). Proteomic data confirmed the activation of a pro-inflammatory signature associated to TLR4 and nuclear factor kappa B (NFkB) activation. Moreover, mH2A1.1-OE proteomic profile identified several upregulated proteins associated to DNA and histones hypermethylation, including S-adenosylhomocysteine hydrolase, a strong inhibitor of DNA methyltransferase and of the methyl donor S-adenosyl-methionine (SAM). HPLC analysis confirmed higher SAM/SAH ratio along with a metabolic reprogramming. Interestingly, an increased LDHA nuclear localization was detected both in mH2A1.1-OE cells and MDS-MSCs, probably depending on MSC inflammatory phenotype. Finally, coculturing healthy mH2A1.1-OE MSCs with CD34+ cells, we found a significant reduction in the number of CD34+ cells, which was reflected in a decreased number of colony forming units (CFU-Cs). These results suggest a key role of mH2A1.1 in driving the crosstalk between epigenetic signaling, inflammation, and cell metabolism networks in MDS-MSCs.
Collapse
Affiliation(s)
- C Giallongo
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | - I Dulcamare
- Division of Hematology, AOU Policlinico, Catania, Italy
| | - S Giallongo
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy.
| | - A Duminuco
- Division of Hematology, AOU Policlinico, Catania, Italy
| | - D Pieragostino
- Department of Innovative Technologies and Medicine & Odontoiatry, University G. D'Annunzio, Chieti-Pescara, Italy
- Analytical Biochemistry and Proteomics Laboratory, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - M C Cufaro
- Department of Innovative Technologies and Medicine & Odontoiatry, University G. D'Annunzio, Chieti-Pescara, Italy
- Analytical Biochemistry and Proteomics Laboratory, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - A M Amorini
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - G Lazzarino
- Departmental Faculty of Medicine and Surgery, UniCamillus-Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - A Romano
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - N Parrinello
- Division of Hematology, AOU Policlinico, Catania, Italy
| | - M Di Rosa
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - G Broggi
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | - R Caltabiano
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | - M Caraglia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
- Laboratory of Precision and Molecular Oncology, Biogem Scarl, Institute of Genetic Research, Ariano Irpino, Italy
| | - M Scrima
- Laboratory of Precision and Molecular Oncology, Biogem Scarl, Institute of Genetic Research, Ariano Irpino, Italy
| | - L S Pasquale
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
- Laboratory of Precision and Molecular Oncology, Biogem Scarl, Institute of Genetic Research, Ariano Irpino, Italy
| | - M S Tathode
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
- Laboratory of Precision and Molecular Oncology, Biogem Scarl, Institute of Genetic Research, Ariano Irpino, Italy
| | - G Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| | - R Motterlini
- Faculty of Health, University Paris Est Créteil, INSERM, IMRB, Créteil, France
| | - F Di Raimondo
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - D Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - G A Palumbo
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| |
Collapse
|
8
|
Liesveld J, Galipeau J. In Vitro Insights Into the Influence of Marrow Mesodermal/Mesenchymal Progenitor Cells on Acute Myelogenous Leukemia and Myelodysplastic Syndromes. Stem Cells 2023; 41:823-836. [PMID: 37348128 DOI: 10.1093/stmcls/sxad050] [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: 03/20/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023]
Abstract
The study of marrow-resident mesodermal progenitors can provide important insight into their role in influencing normal and aberrant hematopoiesis as occurs in acute myelogenous leukemia (AML) and myelodysplastic syndromes (MDS). In addition, the chemokine competency of these cells provides links to the inflammatory milieu of the marrow microenvironment with additional implications for normal and malignant hematopoiesis. While in vivo studies have elucidated the structure and function of the marrow niche in murine genetic models, corollary human studies have not been feasible, and thus the use of culture-adapted mesodermal cells has provided insights into the role these rare endogenous niche cells play in physiologic, malignant, and inflammatory states. This review focuses on culture-adapted human mesenchymal stem/stromal cells (MSCs) as they have been utilized in understanding their influence in AML and MDS as well as on their chemokine-mediated responses to myeloid malignancies, injury, and inflammation. Such studies have intrinsic limitations but have provided mechanistic insights and clues regarding novel druggable targets.
Collapse
Affiliation(s)
- Jane Liesveld
- Department of Medicine, James P. Wilmot Cancer Institute, University of Rochester, Rochester, NY, USA
| | - Jaques Galipeau
- University of Wisconsin School of Medicine and Public Health, University of Wisconsin in Madison, Madison, WI, USA
| |
Collapse
|
9
|
Koerber RM, Schneider RK, Pritchard JE, Teichmann LL, Schumacher U, Brossart P, Gütgemann I. Nestin expression in osteocytes following myeloablation and during bone marrow metastasis. Br J Haematol 2023; 200:643-651. [PMID: 36382360 DOI: 10.1111/bjh.18563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/19/2022] [Accepted: 11/02/2022] [Indexed: 11/18/2022]
Abstract
Nestin is an intermediate filament protein, which was originally detected in neuroepithelial stem cells. Besides its use as a phenotypic marker of mesenchymal stem cells in the hematopoeitic stem cell niche, the functional interpretation of nestin+ cells remains elusive. We investigated the cellular expression of nestin in bone marrow trephine biopsies of MPN patients, following myeloablation at a stage of hypocellularity during early regeneration. Here, nestin is highly expressed in mature osteocytes, arteriolar endothelial and perivascular cells and small capillaries within the bone marrow space, but not in sinusoid lining cells. This is in stark contrast to nestin expression pattern in myeloproliferative neoplasms that show hypercellularity due to oncogenic driver mutations. Here, nestin is expressed exclusively in endothelial cells of arterioles, but not in osteocytes or small capillaries. Thus, the pattern of nestin expression following myeloablation inversely correlates with cellularity in the bone marrow. This nestin expression pattern is mimicking early postnatal transcriptional programming during bone marrow development. We show that nestin expression in osteocytes occurs across different species following transplant and also in bone marrow metastasis.
Collapse
Affiliation(s)
- Ruth-Miriam Koerber
- Department of Medicine III, University Hospital Bonn, Bonn, Germany.,Mildred Scheel School of Oncology, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Rebekka K Schneider
- Department of Cell Biology, Institute for Biomedical Engineering, Aachen, Germany
| | | | - Lino L Teichmann
- Department of Medicine III, University Hospital Bonn, Bonn, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Brossart
- Department of Medicine III, University Hospital Bonn, Bonn, Germany
| | - Ines Gütgemann
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
10
|
Remodeled CD146 +CD271 + Bone Marrow Mesenchymal Stem Cells from Patients with Polycythemia Vera Exhibit Altered Hematopoietic Supportive Activity. Stem Cell Rev Rep 2023; 19:406-416. [PMID: 36018465 DOI: 10.1007/s12015-022-10427-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 02/07/2023]
Abstract
An essential component of the hematopoietic microenvironment, bone marrow mesenchymal stem cells (BM-MSCs) play an important role in the homeostasis and pathogenesis of the hematopoietic system by regulating the fate of hematopoietic stem cells (HSCs). Previous studies revealed that BM-MSCs were functionally remodeled by malignant cells in leukemia. However, the alterations in BM-MSCs in polycythemia vera (PV) and their effects on HSCs still need to be elucidated. Our results demonstrated that although BM-MSCs from PV patients shared similar surface markers with those from healthy donors, they exhibited enhanced proliferation, decreased senescence, and abnormal osteogenic differentiation capacities. The CD146+CD271+ BM-MSC subpopulation, which is considered to give rise to typical cultured BM-MSCs and form bone and the hematopoietic stroma, was then sorted. Compared with those from healthy donors, CD146+CD271+ BM-MSCs from PV patients showed an impaired mesensphere formation capacity and abnormal differentiation toward osteogenic lineages. In addition, CD146+CD271+ PV BM-MSCs showed altered hematopoietic supportive activity when cocultured with cord blood CD34+ cells. Our study suggested that remodeled CD146+CD271+ BM-MSCs might contribute to the pathogenesis of PV, a finding that will shed light on potential therapeutic strategies for PV.
Collapse
|
11
|
Marson RF, Regner AP, da Silva Meirelles L. Mesenchymal "stem" cells, or facilitators for the development of regenerative macrophages? Pericytes at the interface of wound healing. Front Cell Dev Biol 2023; 11:1148121. [PMID: 36936686 PMCID: PMC10017474 DOI: 10.3389/fcell.2023.1148121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Cultured mesenchymal stromal cells are among the most used cells in clinical trials. Currently, their potential benefits include provision of mature cell types through differentiation, and secretion of various types of paracrine signaling molecules. Even though research on these cells has spanned some decades now, surprisingly, their therapeutic potential has not been fully translated into clinical practice yet, which calls for further understanding of their intrinsic nature and modes of action. In this review, after discussing pieces of evidence that suggest that some perivascular cells may exhibit mesenchymal stem cell characteristics in vivo, we examine the possibility that subpopulations of perivascular and/or adventitial cells activated after tissue injury behave as MSCs and contribute to the resolution of tissue injury by providing cues for the development of regenerative macrophages at injured sites. Under this perspective, an important contribution of cultured MSCs (or their acellular products, such as extracellular vesicles) used in cell therapies would be to instigate the development of M2-like macrophages that support the tissue repair process.
Collapse
Affiliation(s)
- Renan Fava Marson
- Graduate Program in Cellular and Molecular Biology Applied to Health—PPGBioSaúde, Lutheran University of Brazil, Canoas, Brazil
| | - Andrea Pereira Regner
- Graduate Program in Cellular and Molecular Biology Applied to Health—PPGBioSaúde, Lutheran University of Brazil, Canoas, Brazil
- School of Medicine, Lutheran University of Brazil, Canoas, Brazil
| | - Lindolfo da Silva Meirelles
- Graduate Program in Cellular and Molecular Biology Applied to Health—PPGBioSaúde, Lutheran University of Brazil, Canoas, Brazil
- School of Medicine, Lutheran University of Brazil, Canoas, Brazil
- *Correspondence: Lindolfo da Silva Meirelles, ,
| |
Collapse
|
12
|
Qu B, Han X, Zhao L, Zhang F, Gao Q. Relationship of HIF‑1α expression with apoptosis and cell cycle in bone marrow mesenchymal stem cells from patients with myelodysplastic syndrome. Mol Med Rep 2022; 26:239. [PMID: 35642674 PMCID: PMC9185697 DOI: 10.3892/mmr.2022.12755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 02/21/2022] [Indexed: 11/09/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a group of abnormal clonal disorders with ineffective hematopoiesis, which are incurable with conventional therapy. Of note, MDS features an abnormal bone marrow microenvironment, which is related to its incidence. The hypoxia-inducible factor-1α (HIF-1α) transcriptional signature is generally activated in bone marrow stem/progenitor cells of patients with MDS. To analyze the expression of HIF-1α in bone marrow mesenchymal stem cells (BM-MSCs) and the apoptosis and cell cycle features associated with the disease, BM-MSCs were obtained from 40 patients with a definitive diagnosis of MDS and 20 subjects with hemocytopenia but a negative diagnosis of MDS as a control group. Reverse transcription-quantitative PCR and western blot analyses were used to measure HIF-1α expression in cells from the two groups and apoptosis and cell cycle were also analyzed and compared between the groups using flow cytometry assays. BM-MSCs from both the control group and the MDS group exhibited a fibroblast-like morphology, had similar growth cycles and were difficult to passage stably. It was observed that BM-MSCs from the MDS group had significantly higher HIF-1α expression levels than the control group (P<0.05). Furthermore, the BM-MSCs from the MDS group had a higher proportion of cells in early apoptosis (5.22±1.34 vs. 2.04±0.08%; P<0.0001) and late apoptosis (3.38±0.43 vs. 1.23±0.11%; P<0.01) and exhibited cell cycle arrest. This may be a noteworthy aspect of the pathogenesis of MDS and may be related to high HIF-1α expression under a hypoxic state in the bone marrow microenvironment. Furthermore, the expression of HIF-1α in bone marrow tissue sections from patients with MDS in the International Prognostic Scoring System (IPSS) lower-risk group was higher than that from patients with MDS in the IPSS high-risk group. These results revealed the role of HIF-1α as a central pathobiology mediator of MDS and an effective therapeutic target for a broad spectrum of patients with MDS, particularly for patients in the lower-risk group.
Collapse
Affiliation(s)
- Beibei Qu
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Xiuhua Han
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Lan Zhao
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Feifei Zhang
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Qingmei Gao
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| |
Collapse
|
13
|
Menter T, Tzankov A. Tumor Microenvironment in Acute Myeloid Leukemia: Adjusting Niches. Front Immunol 2022; 13:811144. [PMID: 35273598 PMCID: PMC8901718 DOI: 10.3389/fimmu.2022.811144] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/03/2022] [Indexed: 12/19/2022] Open
Abstract
Acute myeloid leukemias (AML) comprise a wide array of different entities, which have in common a rapid expansion of myeloid blast cells leading to displacement of normal hematopoietic cells and also disruption of the microenvironment in the bone marrow niches. Based on an insight into the complex cellular interactions in the bone marrow niches in non-neoplastic conditions in general, this review delineates the complex relationship between leukemic cells and reactive cells of the tumor microenvironment (TME) in AML. A special focus is directed on niche cells and various T-cell subsets as these also provide a potential therapeutic rationale considering e.g. immunomodulation. The TME of AML on the one hand plays a vital role for sustaining and promoting leukemogenesis but - on the other hand - it also has adverse effects on abnormal blasts developing into overt leukemia hindering their proliferation and potentially removing such cells. Thus, leukemic cells need to and develop strategies in order to manipulate the TME. Interference with those strategies might be of particular therapeutic potential since mechanisms of resistance related to tumor cell plasticity do not apply to it.
Collapse
Affiliation(s)
- Thomas Menter
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alexandar Tzankov
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
14
|
Omatsu Y, Higaki K, Nagasawa T. Cellular Niches for Hematopoietic Stem Cells and Lympho-Hematopoiesis in Bone Marrow During Homeostasis and Blood Cancers. Curr Top Microbiol Immunol 2021; 434:33-54. [PMID: 34850281 DOI: 10.1007/978-3-030-86016-5_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Most types of blood cells, including immune cells are generated from hematopoietic stem cells (HSCs) within bone marrow in the adult. Most HSCs are in contact with and require the special microenvironment known as a niche for their maintenance. It has been thought that HSC niches comprise various types of support cells that provide critical signals, including cytokines and extracellular matrix for HSC regulation. However, among these cells, several lines of evidence have demonstrated that the population of bone marrow-specific mesenchymal stem cells, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells, which overlap strongly with leptin receptor-expressing (LepR+) cells, is the major cellular component of HSC niches. CAR/LepR+ cells give rise to most adipocytes and osteoblasts in adult bone marrow and express much higher levels of HSC niche factors, including cytokines CXCL12 and stem cell factor (SCF), which are essential for HSC maintenance, and transcription factors Foxc1 and Ebf3, which are essential for the formation and maintenance of HSC niches than other types of cells. CAR/LepR+ cells are present in human bone marrow, undergo fibrotic expansion, and have reduced expression of HSC niche factors in hematopoietic malignancies.
Collapse
Affiliation(s)
- Yoshiki Omatsu
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Kei Higaki
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, 565-0871, Osaka, Japan.
| |
Collapse
|
15
|
Cagnan I, Keles M, Keskus AG, Tombaz M, Sahan OB, Aerts-Kaya F, Uckan-Cetinkaya D, Konu O, Gunel-Ozcan A. Global miRNA expression of bone marrow mesenchymal stem/stromal cells derived from Fanconi anemia patients. Hum Cell 2021; 35:111-124. [PMID: 34792755 DOI: 10.1007/s13577-021-00626-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 09/26/2021] [Indexed: 11/24/2022]
Abstract
Fanconi anemia (FA) is a rare genetic disorder characterized by genomic instability, developmental defects, and bone marrow (BM) failure. Hematopoietic stem cells (HSCs) in BM interact with the mesenchymal stem/stromal cells (MSCs); and this partly sustains the tissue homeostasis. MicroRNAs (miRNAs) can play a critical role during these interactions possibly via paracrine mechanisms. This is the first study addressing the miRNA profile of FA BM-MSCs obtained before and after BM transplantation (preBMT and postBMT, respectively). Non-coding RNA expression profiling and quality control analyses were performed in Donors (n = 13), FA preBMT (n = 11), and FA postBMT (n = 6) BM-MSCs using GeneChip miRNA 2.0 Array. Six Donor-FA preBMT pairs were used to identify a differentially expressed miRNA expression signature containing 50 miRNAs, which exhibited a strong correlation with the signature obtained from unpaired samples. Five miRNAs (hsa-miR-146a-5p, hsa-miR-148b-3p, hsa-miR-187-3p, hsa-miR-196b-5p, and hsa-miR-25-3p) significantly downregulated in both the paired and unpaired analyses were used to generate the BM-MSCs' miRNA-BM mononuclear mRNA networks upon integration of a public dataset (GSE16334; studying Donor versus FA samples). Functionally enriched KEGG pathways included cellular senescence, miRNAs, and pathways in cancer. Here, we showed that hsa-miR-146a-5p and hsa-miR-874-3p were rescued upon BMT (n = 3 triplets). The decrease in miR-146a-5p was also validated using RT-qPCR and emerged as a strong candidate as a modulator of BM mRNAs in FA patients.
Collapse
Affiliation(s)
- Ilgin Cagnan
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Center for Stem Cell Research and Development, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.,Department of Biological Sciences, Faculty of Arts and Sciences, Eastern Mediterranean University, 99628, Famagusta, North Cyprus, via Mersin-10, Turkey
| | - Mustafa Keles
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Center for Stem Cell Research and Development, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.,Center for Stem Cell Research and Development, PEDI-STEM, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Ayse Gokce Keskus
- Interdisciplinary Neuroscience Program, Bilkent University, Ankara, Turkey
| | - Melike Tombaz
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Ozge Burcu Sahan
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Center for Stem Cell Research and Development, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.,Center for Stem Cell Research and Development, PEDI-STEM, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Fatima Aerts-Kaya
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Center for Stem Cell Research and Development, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.,Center for Stem Cell Research and Development, PEDI-STEM, Hacettepe University, 06100, Sihhiye, Ankara, Turkey
| | - Duygu Uckan-Cetinkaya
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Center for Stem Cell Research and Development, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.,Center for Stem Cell Research and Development, PEDI-STEM, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.,Department of Pediatrics, Division of Bone Marrow Transplantation Unit, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ozlen Konu
- Interdisciplinary Neuroscience Program, Bilkent University, Ankara, Turkey. .,Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey.
| | - Aysen Gunel-Ozcan
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Center for Stem Cell Research and Development, Hacettepe University, 06100, Sihhiye, Ankara, Turkey. .,Center for Stem Cell Research and Development, PEDI-STEM, Hacettepe University, 06100, Sihhiye, Ankara, Turkey.
| |
Collapse
|
16
|
Omatsu Y, Nagasawa T. Identification of microenvironmental niches for hematopoietic stem cells and lymphoid progenitors-bone marrow fibroblastic reticular cells with salient features. Int Immunol 2021; 33:821-826. [PMID: 34668936 DOI: 10.1093/intimm/dxab092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 10/19/2021] [Indexed: 12/14/2022] Open
Abstract
Most lineages of blood cells, including immune cells are generated from hematopoietic stem cells (HSCs) in bone marrow throughout adult life. Since HSCs cannot expand on their own, they require and contact the special microenvironments, termed niches for their maintenance. HSC niches comprise supportive cells that provide adjacent HSCs with critical signals, including cytokines. Although bone marrow microenvironments have been thought to be complex, recent studies have demonstrated that the bone marrow-specific population of fibroblastic reticular cells with long processes, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells, which overlap strongly with leptin receptor (LepR)-expressing (LepR +) cells, is the major cellular component of niches for HSCs and lymphoid progenitors. CAR cells have salient features, expressing much higher levels of critical HSC niche factors than any other cell populations and function as self-renewing mesenchymal stem cells. Human counterpart of CAR cells is present and affected in diseases, including leukemia. Foxl1 + telocytes recently identified as the niche for intestinal stem cells share some features with CAR cells, suggesting that CAR cells are prototypical fibroblastic reticular cells creating niche for long-lived cells, including tissue stem cells and memory lymphocytes. These findings provided the basis for future mechanistic studies on the crosstalk between hematopoietic cells and microenvironments in both health and disease.
Collapse
Affiliation(s)
- Yoshiki Omatsu
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
17
|
The Impact of Sedentary Lifestyle, High-fat Diet, Tobacco Smoke, and Alcohol Intake on the Hematopoietic Stem Cell Niches. Hemasphere 2021; 5:e615. [PMID: 34291194 PMCID: PMC8288907 DOI: 10.1097/hs9.0000000000000615] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/07/2021] [Indexed: 11/25/2022] Open
Abstract
Hematopoietic stem and progenitor cells maintain hematopoiesis throughout life by generating all major blood cell lineages through the process of self-renewal and differentiation. In adult mammals, hematopoietic stem cells (HSCs) primarily reside in the bone marrow (BM) at special microenvironments called “niches.” Niches are thought to extrinsically orchestrate the HSC fate including their quiescence and proliferation. Insight into the HSC niches mainly comes from studies in mice using surface marker identification and imaging to visualize HSC localization and association with niche cells. The advantage of mouse models is the possibility to study the 3-dimensional BM architecture and cell interactions in an intact traceable system. However, this may not be directly translational to human BM. Sedentary lifestyle, unhealthy diet, excessive alcohol intake, and smoking are all known risk factors for various diseases including hematological disorders and cancer, but how do lifestyle factors impact hematopoiesis and the associated niches? Here, we review current knowledge about the HSC niches and how unhealthy lifestyle may affect it. In addition, we summarize epidemiological data concerning the influence of lifestyle factors on hematological disorders and malignancies.
Collapse
|
18
|
Meza-León B, Gratzinger D, Aguilar-Navarro AG, Juárez-Aguilar FG, Rebel VI, Torlakovic E, Purton LE, Dorantes-Acosta EM, Escobar-Sánchez A, Dick JE, Flores-Figueroa E. Human, mouse, and dog bone marrow show similar mesenchymal stromal cells within a distinctive microenvironment. Exp Hematol 2021; 100:41-51. [PMID: 34228982 DOI: 10.1016/j.exphem.2021.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/22/2021] [Accepted: 06/26/2021] [Indexed: 12/22/2022]
Abstract
Bone marrow stromal cells (BMSCs) are a key part of the hematopoietic niche. Mouse and human BMSCs are recognized by different markers (LepR and NGFR/CD271, respectively). However, there has not been a detailed in situ comparison of both populations within the hematopoietic microenvironment. Moreover, dog BMSCs have not been characterized in situ by any of those markers. We conducted a systematic histopathological comparison of mouse, human, and dog BMSCs within their bone marrow architecture and microenvironment. Human and dog CD271+ BMSCs had a morphology, frequency, and distribution within trabecular bone marrow similar to those of mouse LepR+ BMSCs. However, mouse bone marrow had higher cellularity and megakaryocyte content. In conclusion, highly comparable bone marrow mesenchymal stromal cell distribution among the three species establishes the validity of using mouse and dog as a surrogate experimental model of hematopoietic stem cell-BMSC interactions. However, the distinct differences in adipocyte and megakaryocyte microenvironment content of mouse bone marrow and how they might influence hematopoietic stem cell interactions as compared with humans require further study.
Collapse
Affiliation(s)
- Berenice Meza-León
- Unidad de Investigación Médica en Enfermedades Oncológicas. Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, México
| | - Dita Gratzinger
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Alicia G Aguilar-Navarro
- Unidad de Investigación Médica en Enfermedades Oncológicas. Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, México
| | - Fany G Juárez-Aguilar
- Departamento de Patología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, México
| | - Vivienne I Rebel
- Department of Cell Systems & Anatomy, The University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Emina Torlakovic
- Saskatchewan Health Authority (SHA), Saskatoon, Saskatchewan, Canada; University of Saskatchewan, Saskatchewan, Canada
| | - Louise E Purton
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia; Department of Medicine, University of Melbourne, Parkville, VIC, Australia
| | - Elisa M Dorantes-Acosta
- Biobanco de Investigación en Células Leucémicas, Hospital Infantil de México Federico Gómez, Mexico City, México
| | | | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Eugenia Flores-Figueroa
- Unidad de Investigación Médica en Enfermedades Oncológicas. Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, México; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
19
|
Barreto-Duran E, Mejia-Cruz CC, Jaramillo-Garcia LF, Leal-Garcia E, Barreto-Prieto A, Rodriguez-Pardo VM. 3D Multicellular Spheroid for the Study of Human Hematopoietic Stem Cells: Synergistic Effect Between Oxygen Levels, Mesenchymal Stromal Cells and Endothelial Cells. J Blood Med 2021; 12:517-528. [PMID: 34234608 PMCID: PMC8256312 DOI: 10.2147/jbm.s305319] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/27/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction The human bone marrow microenvironment is composed of biological, chemical and physical factors that act in a synergistic way to modulate hematopoietic stem cell biology, such as mesenchymal stromal cells (MSCs), endothelial cells (ECs) and low oxygen levels; however, it is difficult to mimic this human microenvironment in vitro. Methods In this work, we developed 3D multicellular spheroid (3D-MS) for the study of human hematopoietic stem cells (HSCs) with some components of perivascular niche. HSCs were isolated from umbilical cord blood, MSCs were isolated from human bone marrow and a microvasculature EC line (CC-2811, Lonza®) was used. For the formation of a 3D structure, a magnetic levitation culture system was used. Cultures were maintained in 21%, 3% and 1% O2 for 15 days. Culture volume, sphericity index and cell viability were determined. Also, human HSC proliferation, phenotype and production of reactive oxygen species were evaluated. Results After 15 days, 3D-MS exhibited viability greater than 80%. Histology results showed structures without necrotic centers, and higher cellular proliferation with 3% O2. An increase in the expression of the CD34 antigen and other hematopoietic antigens were observed to 1% O2 with MSCs plus ECs and low ROS levels. Conclusion These findings suggest that 3D-MS formed by MSCs, ECs and HSCs exposed to low concentrations of oxygen (1–3% O2) modulate human HSC behavior and mimics some features of the perivascular niche, which could reduce the use of animal models and deepen the relationship between the microenvironment of HSC and human hematological diseases development.
Collapse
Affiliation(s)
- Emilia Barreto-Duran
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia (South America)
| | - Claudia Camila Mejia-Cruz
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia (South America)
| | - Luis Fernando Jaramillo-Garcia
- Departamento de Patología, Facultad de Medicina, Pontificia Universidad Javeriana., Hospital Universitario San Ignacio, Bogotá D.C., Colombia (South America)
| | - Efrain Leal-Garcia
- Departamento de Ortopedia y Traumatología, Facultad de Medicina, Pontificia Universidad Javeriana., Hospital Universitario San Ignacio, Bogotá D.C., Colombia (South America)
| | - Alfonso Barreto-Prieto
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia (South America)
| | - Viviana Marcela Rodriguez-Pardo
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia (South America)
| |
Collapse
|
20
|
Torres-Barrera P, Mayani H, Chávez-González A. Understanding the hematopoietic microenvironment in chronic myeloid leukemia: A concise review. Curr Res Transl Med 2021; 69:103295. [PMID: 33962119 DOI: 10.1016/j.retram.2021.103295] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 04/04/2021] [Accepted: 04/13/2021] [Indexed: 12/01/2022]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disease that results from the BCR-ABL gene-induced transformation of a primitive hematopoietic cell. This disease has been extensively studied, and, as a result, a very effective therapy has been developed: the tyrosine kinase inhibitors. Although, there is a significant knowledge about the intrinsic biology of CML cells, alterations in their bone marrow microenvironment are not yet completely understood. In this concise review, we summarized recent findings on the composition and function of the bone marrow microenvironment in CML, and their importance in the progression of the disease and treatment resistance.
Collapse
Affiliation(s)
- P Torres-Barrera
- Laboratorio de Células Troncales Leucémicas, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN Siglo XXI, Instituto Mexicano del Seguro Social, México; Posgrado en Ciencias Biológicas, UNAM, México
| | - H Mayani
- Laboratorio de Células Troncales Hematopoyéticas, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN Siglo XXI, Instituto Mexicano del Seguro Social, México
| | - A Chávez-González
- Laboratorio de Células Troncales Leucémicas, Unidad de Investigación Médica en Enfermedades Oncológicas, CMN Siglo XXI, Instituto Mexicano del Seguro Social, México.
| |
Collapse
|
21
|
Kristensen HB, Andersen TL, Patriarca A, Kallenbach K, MacDonald B, Sikjaer T, Ejersted C, Delaisse JM. Human hematopoietic microenvironments. PLoS One 2021; 16:e0250081. [PMID: 33878141 PMCID: PMC8057613 DOI: 10.1371/journal.pone.0250081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/30/2021] [Indexed: 11/18/2022] Open
Abstract
Dormancy of hematopoietic stem cells and formation of progenitors are directed by signals that come from the bone marrow microenvironment. Considerable knowledge has been gained on the murine hematopoietic stem cell microenvironment, while less so on the murine progenitor microenvironment and even less so on these microenvironments in humans. Characterization of these microenvironments is decisive for understanding hematopoiesis and finding new treatment modalities against bone marrow malignancies in the clinic. However, it is equally challenging, because hematopoietic stem cells are difficult to detect in the complex bone marrow landscape. In the present study we are characterizing the human hematopoietic stem cell and progenitor microenvironment. We obtained three adjacent bone marrow sections from ten healthy volunteers. One was used to identify a population of CD34+/CD38- “hematopoietic stem cells and multipotent progenitors” and a population of CD34+/CD38+ “progenitors” based on immunofluorescence pattern/intensity and cellular morphology. The other two were immunostained respectively for CD34/CD56 and for CD34/SMA. Using the combined information we performed a non-computer-assisted quantification of nine bone marrow components (adipocytes, megakaryocytes, bone surfaces, four different vessel types (arteries, capillaries, sinusoids and collecting sinuses), other “hematopoietic stem cells and multipotent progenitors” and other “progenitors”) within 30 μm of “hematopoietic stem cells and multipotent progenitors”, “progenitors”, and “random cell profiles”. We show that the microenvironment of the “hematopoietic stem cells and multipotent progenitors” is significantly enriched in sinusoids and megakaryocytes, while the microenvironment of the “progenitors” is significantly enriched in capillaries, other “progenitors”, bone surfaces and arteries.
Collapse
Affiliation(s)
- Helene Bjoerg Kristensen
- Department of Clinical Cell Biology, Institute of Regional Health Science, University of Southern Denmark, Lillebaelt Hospital, Vejle, Denmark
- Department of Pathology, Clinical Cell Biology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research and Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- * E-mail:
| | - Thomas Levin Andersen
- Department of Clinical Cell Biology, Institute of Regional Health Science, University of Southern Denmark, Lillebaelt Hospital, Vejle, Denmark
- Department of Pathology, Clinical Cell Biology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research and Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark
| | - Andrea Patriarca
- Division of Hematology, Department of Oncology, Hospital "Maggiore della Carità", Novara, Italy
| | - Klaus Kallenbach
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Birgit MacDonald
- Department of Clinical Cell Biology, Institute of Regional Health Science, University of Southern Denmark, Lillebaelt Hospital, Vejle, Denmark
| | - Tanja Sikjaer
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Charlotte Ejersted
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Jean-Marie Delaisse
- Department of Clinical Cell Biology, Institute of Regional Health Science, University of Southern Denmark, Lillebaelt Hospital, Vejle, Denmark
- Department of Pathology, Clinical Cell Biology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research and Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
22
|
Aoki K, Kurashige M, Ichii M, Higaki K, Sugiyama T, Kaito T, Ando W, Sugano N, Sakai T, Shibayama H, Takaori-Kondo A, Morii E, Kanakura Y, Nagasawa T. Identification of CXCL12-abundant reticular cells in human adult bone marrow. Br J Haematol 2021; 193:659-668. [PMID: 33837967 PMCID: PMC8252541 DOI: 10.1111/bjh.17396] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/16/2021] [Indexed: 12/14/2022]
Abstract
A population of mesenchymal stem cells, termed CXC chemokine ligand (CXCL)12‐abundant reticular (CAR) cells or leptin receptor‐expressing cells, are the major cellular component of niches for haematopoietic stem cells (HSCs) in murine bone marrow. CAR cells are characterized by several salient features, including much higher expression of CXCL12, stem cell factor (SCF), forkhead box C1 (FOXC1) and early B‐cell factor 3 (EBF3), which are essential for HSC maintenance, than other cells. However, the human counterpart of CAR cells has not been fully described. Here, we show the presence of cells expressing much higher CXCL12 than other cells in human adult bone marrow using a flow cytometry‐based in situ technique that enables high‐throughput detection of mRNA at single‐cell resolution. Most CXCL12hi cells expressed high levels of SCF, FOXC1 and EBF3 and had the potential to differentiate into adipocytes and osteoblasts. Histologically, the nuclei of CXCL12hi cells were identified and quantified by EBF3 expression in fixed marrow sections. CXCL12hi cells sorted from residual bone marrow aspirates of chronic myeloid leukaemia patients expressed reduced levels of CXCL12, SCF, FOXC1 and EBF3 in correlation with increased leukaemic burden. Together, we identified the human counterpart of CAR cells, enabling the evaluation of their alterations in various haematological disorders by flow cytometric and histological analyses.
Collapse
Affiliation(s)
- Kazunari Aoki
- Laboratory of Stem Cell Biology and Developmental Immunology, Immunology Frontier Research Center, World Premier International Research Center (WPI), Graduate School of Frontier Biosciences, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Laboratory of Stem Cell Genetics, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Masako Kurashige
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Michiko Ichii
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kei Higaki
- Laboratory of Stem Cell Biology and Developmental Immunology, Immunology Frontier Research Center, World Premier International Research Center (WPI), Graduate School of Frontier Biosciences, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tatsuki Sugiyama
- Laboratory of Stem Cell Biology and Developmental Immunology, Immunology Frontier Research Center, World Premier International Research Center (WPI), Graduate School of Frontier Biosciences, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takashi Kaito
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Wataru Ando
- Department of Orthopedic Medical Engineering, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Nobuhiko Sugano
- Department of Orthopedic Medical Engineering, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takashi Sakai
- Department of Orthopedic Surgery, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Hirohiko Shibayama
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
| | | | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eiichi Morii
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yuzuru Kanakura
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan.,Sumitomo Hospital, Osaka, Japan
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology and Developmental Immunology, Immunology Frontier Research Center, World Premier International Research Center (WPI), Graduate School of Frontier Biosciences, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
23
|
Aging of Bone Marrow Mesenchymal Stromal Cells: Hematopoiesis Disturbances and Potential Role in the Development of Hematologic Cancers. Cancers (Basel) 2020; 13:cancers13010068. [PMID: 33383723 PMCID: PMC7794884 DOI: 10.3390/cancers13010068] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/16/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary As for many other cancers, the risk of developing hematologic malignancies increases considerably as people age. In recent years, a growing number of studies have highlighted the influence of the aging microenvironment on hematopoiesis and tumor progression. Mesenchymal stromal cells are a major player in intercellular communication inside the bone marrow microenvironment involved in hematopoiesis support. With aging, their functions may be altered, leading to hematopoiesis disturbances which can lead to hematologic cancers. A good understanding of the mechanisms involved in mesenchymal stem cell aging and the consequences on hematopoiesis and tumor progression is therefore necessary for a better comprehension of hematologic malignancies and for the development of therapeutic approaches. Abstract Aging of bone marrow is a complex process that is involved in the development of many diseases, including hematologic cancers. The results obtained in this field of research, year after year, underline the important role of cross-talk between hematopoietic stem cells and their close environment. In bone marrow, mesenchymal stromal cells (MSCs) are a major player in cell-to-cell communication, presenting a wide range of functionalities, sometimes opposite, depending on the environmental conditions. Although these cells are actively studied for their therapeutic properties, their role in tumor progression remains unclear. One of the reasons for this is that the aging of MSCs has a direct impact on their behavior and on hematopoiesis. In addition, tumor progression is accompanied by dynamic remodeling of the bone marrow niche that may interfere with MSC functions. The present review presents the main features of MSC senescence in bone marrow and their implications in hematologic cancer progression.
Collapse
|
24
|
Panvini FM, Pacini S, Montali M, Barachini S, Mazzoni S, Morganti R, Ciancia EM, Carnicelli V, Petrini M. High NESTIN Expression Marks the Endosteal Capillary Network in Human Bone Marrow. Front Cell Dev Biol 2020; 8:596452. [PMID: 33364234 PMCID: PMC7753038 DOI: 10.3389/fcell.2020.596452] [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: 08/19/2020] [Accepted: 11/11/2020] [Indexed: 12/20/2022] Open
Abstract
Hematopoiesis is hosted, supported and regulated by a special bone marrow (BM) microenvironment known as "niche." BM niches have been classified based on micro-anatomic distance from the bone surface into "endosteal" and "central" niches. Whilst different blood vessels have been found in both BM niches in mice, our knowledge of the human BM architecture is much more limited. Here, we have used a combination of markers including NESTIN, CD146, and αSMA labeling different blood vessels in benign human BM. Applying immunohistochemical/immunofluorescence techniques on BM trephines and performing image analysis on almost 300 microphotographs, we detected high NESTIN expression in BM endothelial cells (BMECs) of small arteries (A) and endosteal arterioles (EA), and also in very small vessels we named NESTIN+ capillary-like tubes (NCLTs), not surrounded by sub-endothelial perivascular cells that occasionally reported low levels of NESTIN expression. Statistically, NCLTs were detected within 40 μm from bone trabecula, frequently found in direct contact to the bone line and spatially correlated with hematopoietic stem/progenitor cells. Our results support the expression of NESTIN in human BMECs of EA and A in accordance with the updated classification of murine BM micro-vessels. NCLTs for their peculiar characteristics and micro-anatomical localization have been here proposed as transitional vessels possibly involved in regulating human hematopoiesis.
Collapse
Affiliation(s)
- Francesca M. Panvini
- Institute of Life Sciences, Sant’Anna School of Advanced Studies, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Simone Pacini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marina Montali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Serena Barachini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefano Mazzoni
- Department of Translational Research and New Technology in Medicine, University of Pisa, Pisa, Italy
| | - Riccardo Morganti
- Statistical Support to Clinical Trials Department, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Eugenio M. Ciancia
- Department of Pathology, Azienda Ospedsaliero Universitaria Pisana, Pisa, Italy
| | - Vittoria Carnicelli
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Mario Petrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
25
|
Aguilar-Navarro AG, Meza-León B, Gratzinger D, Juárez-Aguilar FG, Chang Q, Ornatsky O, Tsui H, Esquivel-Gómez R, Hernández-Ramírez A, Xie SZ, Dick JE, Flores-Figueroa E. Human Aging Alters the Spatial Organization between CD34+ Hematopoietic Cells and Adipocytes in Bone Marrow. Stem Cell Reports 2020; 15:317-325. [PMID: 32649902 PMCID: PMC7419665 DOI: 10.1016/j.stemcr.2020.06.011] [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: 02/29/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 02/08/2023] Open
Abstract
Age-related clonal hematopoiesis is a major risk factor for myeloid malignancy and myeloid skewing is a hallmark of aging. However, while it is known that non-cell-autonomous components of the microenvironment can also influence this risk, there have been few studies of how the spatial architecture of human bone marrow (BM) changes with aging. Here, we show that BM adiposity increases with age, which correlates with increased density of maturing myeloid cells and CD34+ hematopoietic stem/progenitor cells (HSPCs) and an increased proportion of HSPCs adjacent to adipocytes. However, NGFR+ bone marrow stromal cell (NGFR+ BMSC) density and distance to HSPCs and vessels remained stable. Interestingly, we found that, upon aging, maturing myeloid cell density increases in hematopoietic areas surrounding adipocytes. We propose that increased adjacency to adipocytes in the BM microenvironment may influence myeloid skewing of aging HSPCs, contributing to age-related risk of myeloid malignancies. Aging increases adipose, myeloid, and CD34+ HSPC density in the human bone marrow Human CD34+ HSPC niche is reticular, perivascular, and periadipocytic in aging Aging increases maturing myeloid cell density surrounding adipocytes
Collapse
Affiliation(s)
- Alicia G Aguilar-Navarro
- Unidad de Investigación Médica en Enfermedades Oncológicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Berenice Meza-León
- Unidad de Investigación Médica en Enfermedades Oncológicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Dita Gratzinger
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Fany G Juárez-Aguilar
- Departamento de Patología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Qing Chang
- Fluidigm Canada Inc., Markham, ON, Canada
| | | | - Hubert Tsui
- Division of Hematopathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Ricardo Esquivel-Gómez
- División de Ortopedia, Hospital de Traumatología y Ortopedia Lomas Verdes, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Antonio Hernández-Ramírez
- Unidad de Reemplazo Articular, Hospital de Traumatología y Ortopedia Lomas Verdes, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Stephanie Z Xie
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Eugenia Flores-Figueroa
- Unidad de Investigación Médica en Enfermedades Oncológicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
26
|
|
27
|
Sun T, Ju M, Dai X, Dong H, Gu W, Gao Y, Fu R, Liu X, Huang Y, Liu W, Ch Y, Wang W, Li H, Zhou Y, Shi L, Yang R, Zhang L. Multilevel defects in the hematopoietic niche in essential thrombocythemia. Haematologica 2019; 105:661-673. [PMID: 31289202 PMCID: PMC7049349 DOI: 10.3324/haematol.2018.213686] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 07/05/2019] [Indexed: 12/24/2022] Open
Abstract
The role of the bone marrow niche in essential thrombocythemia (ET) remains unclear. Here, we observed multilevel defects in the hematopoietic niche of patients with JAK2V617F-positive ET, including functional deficiency in mesenchymal stromal cells (MSC), immune imbalance, and sympathetic-nerve damage. Mesenchymal stromal cells from patients with JAK2V617F-positive essential thrombocythemia had a transformed transcriptome. In parallel, they showed enhanced proliferation, decreased apoptosis and senescence, attenuated ability to differentiate into adipocytes and osteocytes, and insufficient support for normal hematopoiesis. Additionally, they were inefficient in suppressing immune responses. For instance, they poorly inhibited proliferation and activation of CD4-positive T cells and the secretion of the inflammatory factor soluble CD40-ligand. They also poorly induced formation of mostly immunosuppressive T-helper 2 cells (Th2) and the secretion of the anti-inflammatory factor interleukin-4 (IL-4). Furthermore, we identified WDR4 as a potent protein with low expression and which was correlated with increased proliferation, reduced senescence and differentiation, and insufficient support for normal hematopoiesis in MSC from patients with JAK2V617F-positive ET. We also observed that loss of WDR4 in MSC cells downregulated the interleukin-6 (IL-6) level through the ERK–GSK3β–CREB signaling based on our in vitro studies. Altogether, our results show that multilevel changes occur in the bone marrow niche of patients with JAK2V617F-positive ET, and low expression of WDR4 in MSC may be critical for inducing hematopoietic related changes.
Collapse
Affiliation(s)
- Ting Sun
- State Key Laboratory of Experimental Hematology.,Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin
| | - Mankai Ju
- State Key Laboratory of Experimental Hematology.,Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin
| | - Xinyue Dai
- State Key Laboratory of Experimental Hematology
| | - Huan Dong
- State Key Laboratory of Experimental Hematology
| | - Wenjing Gu
- State Key Laboratory of Experimental Hematology
| | - Yuchen Gao
- State Key Laboratory of Experimental Hematology
| | - Rongfeng Fu
- State Key Laboratory of Experimental Hematology.,Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin.,Tianjin Laboratory of Blood Disease Gene Therapy.,CAMS Key Laboratory of Gene Therapy for Blood Diseases
| | - Xiaofan Liu
- State Key Laboratory of Experimental Hematology.,Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin.,Tianjin Laboratory of Blood Disease Gene Therapy.,CAMS Key Laboratory of Gene Therapy for Blood Diseases
| | - Yueting Huang
- State Key Laboratory of Experimental Hematology.,Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin.,Tianjin Laboratory of Blood Disease Gene Therapy.,CAMS Key Laboratory of Gene Therapy for Blood Diseases
| | - Wei Liu
- State Key Laboratory of Experimental Hematology.,Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin.,Tianjin Laboratory of Blood Disease Gene Therapy.,CAMS Key Laboratory of Gene Therapy for Blood Diseases
| | - Ying Ch
- State Key Laboratory of Experimental Hematology.,Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin.,Tianjin Laboratory of Blood Disease Gene Therapy.,CAMS Key Laboratory of Gene Therapy for Blood Diseases
| | - Wentian Wang
- State Key Laboratory of Experimental Hematology.,Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin.,Tianjin Laboratory of Blood Disease Gene Therapy.,CAMS Key Laboratory of Gene Therapy for Blood Diseases
| | - Huiyuan Li
- State Key Laboratory of Experimental Hematology.,Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin.,Tianjin Laboratory of Blood Disease Gene Therapy.,CAMS Key Laboratory of Gene Therapy for Blood Diseases
| | - Yuan Zhou
- State Key Laboratory of Experimental Hematology.,Tianjin Laboratory of Blood Disease Gene Therapy.,PUMC Department of Stem Cell and Regenerative Medicine, Tianjin, China
| | - Lihong Shi
- State Key Laboratory of Experimental Hematology .,Tianjin Laboratory of Blood Disease Gene Therapy.,CAMS Center for Stem Cell Medicine.,PUMC Department of Stem Cell and Regenerative Medicine, Tianjin, China
| | - Renchi Yang
- State Key Laboratory of Experimental Hematology .,National Clinical Research Center for Blood Diseases.,Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin.,Tianjin Laboratory of Blood Disease Gene Therapy.,CAMS Key Laboratory of Gene Therapy for Blood Diseases.,CAMS Center for Stem Cell Medicine
| | - Lei Zhang
- State Key Laboratory of Experimental Hematology .,National Clinical Research Center for Blood Diseases.,Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin.,Tianjin Laboratory of Blood Disease Gene Therapy.,CAMS Key Laboratory of Gene Therapy for Blood Diseases.,CAMS Center for Stem Cell Medicine.,PUMC Department of Stem Cell and Regenerative Medicine, Tianjin, China
| |
Collapse
|
28
|
Agarwal P, Isringhausen S, Li H, Paterson AJ, He J, Gomariz Á, Nagasawa T, Nombela-Arrieta C, Bhatia R. Mesenchymal Niche-Specific Expression of Cxcl12 Controls Quiescence of Treatment-Resistant Leukemia Stem Cells. Cell Stem Cell 2019; 24:769-784.e6. [PMID: 30905620 PMCID: PMC6499704 DOI: 10.1016/j.stem.2019.02.018] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 10/01/2018] [Accepted: 02/25/2019] [Indexed: 12/25/2022]
Abstract
Chronic myeloid leukemia (CML) originates in a hematopoietic stem cell (HSC) transformed by the breakpoint cluster region (BCR)-abelson (ABL) oncogene and is effectively treated with tyrosine kinase inhibitors (TKIs). TKIs do not eliminate disease-propagating leukemic stem cells (LSCs), suggesting a deeper understanding of niche-dependent regulation of CML LSCs is required to eradicate disease. Cxcl12 is expressed in bone marrow niches and controls HSC maintenance, and here, we show that targeted deletion of Cxcl12 from mesenchymal stromal cells (MSCs) reduces normal HSC numbers but promotes LSC expansion by increasing self-renewing cell divisions, possibly through enhanced Ezh2 activity. In contrast, endothelial cell-specific Cxcl12 deletion decreases LSC proliferation, suggesting niche-specific effects. During CML development, abnormal clusters of colocalized MSCs and LSCs form but disappear upon Cxcl12 deletion. Moreover, MSC-specific deletion of Cxcl12 increases LSC elimination by TKI treatment. These findings highlight a critical role of niche-specific effects of Cxcl12 expression in maintaining quiescence of TKI-resistant LSC populations.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cell Line, Tumor
- Chemokine CXCL12/genetics
- Chemokine CXCL12/metabolism
- Drug Resistance, Neoplasm
- Enhancer of Zeste Homolog 2 Protein/genetics
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Hematopoietic Stem Cells/physiology
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Mesenchymal Stem Cells/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplastic Stem Cells/physiology
- Organ Specificity
- Protein Kinase Inhibitors/therapeutic use
- Stem Cell Niche/physiology
Collapse
Affiliation(s)
- Puneet Agarwal
- Division of Hematology & Oncology, University of Alabama, Birmingham, Birmingham, AL, USA
| | - Stephan Isringhausen
- Department of Hematology and Oncology, Division of Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Hui Li
- Division of Hematology & Oncology, University of Alabama, Birmingham, Birmingham, AL, USA
| | - Andrew J Paterson
- Division of Hematology & Oncology, University of Alabama, Birmingham, Birmingham, AL, USA
| | - Jianbo He
- Division of Hematology & Oncology, University of Alabama, Birmingham, Birmingham, AL, USA
| | - Álvaro Gomariz
- Department of Hematology and Oncology, Division of Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology & Developmental Immunology, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - César Nombela-Arrieta
- Department of Hematology and Oncology, Division of Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Ravi Bhatia
- Division of Hematology & Oncology, University of Alabama, Birmingham, Birmingham, AL, USA.
| |
Collapse
|
29
|
Abstract
Abstract
Myelodysplastic syndrome (MDS) is characterized by bone marrow failure and a strong propensity for leukemic evolution. Somatic mutations are critical early drivers of the disorder, but the factors enabling the emergence, selection, and subsequent leukemic evolution of these “leukemia-poised” clones remain incompletely understood. Emerging data point at the mesenchymal niche as a critical contributor to disease initiation and evolution. Disrupted inflammatory signaling from niche cells may facilitate the occurrence of somatic mutations, their selection, and subsequent clonal expansion. This review summarizes the current concepts about “niche-facilitated” bone marrow failure and leukemic evolution, their underlying molecular mechanisms, and clinical implications for future innovative therapeutic targeting of the niche in MDS.
Collapse
|
30
|
Petters O, Schmidt C, Henkelmann R, Pieroh P, Hütter G, Marquass B, Aust G, Schulz RM. Single-Stage Preparation of Human Cartilage Grafts Generated from Bone Marrow-Derived CD271 + Mononuclear Cells. Stem Cells Dev 2019; 27:545-555. [PMID: 29482445 DOI: 10.1089/scd.2017.0218] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Due to the limited self-healing capacity of articular cartilage, innovative, regenerative approaches are of particular interest. The use of two-stage procedures utilizing in vitro-expanded mesenchymal stromal cells (MSCs) from various cell sources requires good manufacturing practice-compliant production, a process with high demands on time, staffing, and financial resources. In contrast, one- stage procedures are directly available, but need a safe enrichment of potent MSCs. CD271 is a surface marker known to marking the majority of native MSCs in bone marrow (BM). In this study, the feasibility of generating a single-stage cartilage graft of enriched CD271+ BM-derived mononuclear cells (MNCs) without in vitro monolayer expansion from eight healthy donors was investigated. Cartilage grafts were generated by magnetic-activated cell sorting and separated cells were directly transferred into collagen type I hydrogels, followed by 3D proliferation and differentiation period of CD271+, CD271-, or unseparated MNCs. CD271+ MNCs showed the highest proliferation rate, cell viability, sulfated glycosaminoglycan deposition, and cartilage marker expression compared to the CD271- or unseparated MNC fractions in 3D culture. Analysis according to the minimal criteria of the International Society for Cellular Therapy highlighted a 66.8-fold enrichment of fibroblast colony-forming units in CD271+ MNCs and the only fulfillment of the MSC marker profile compared to unseparated MNCs. In summary, CD271+ MNCs are capable of generating adequate articular cartilage grafts presenting high cell viability and notable chondrogenic matrix deposition in a CE-marked collagen type I hydrogel, which can obviate the need for an initial monolayer expansion.
Collapse
Affiliation(s)
- Oliver Petters
- 1 Centre for Biotechnology and Biomedicine, University of Leipzig , Leipzig, Germany .,2 Department of Orthopedics, Trauma and Plastic Surgery, University of Leipzig , Leipzig, Germany
| | - Christian Schmidt
- 1 Centre for Biotechnology and Biomedicine, University of Leipzig , Leipzig, Germany .,2 Department of Orthopedics, Trauma and Plastic Surgery, University of Leipzig , Leipzig, Germany
| | - Ralf Henkelmann
- 2 Department of Orthopedics, Trauma and Plastic Surgery, University of Leipzig , Leipzig, Germany
| | - Philipp Pieroh
- 2 Department of Orthopedics, Trauma and Plastic Surgery, University of Leipzig , Leipzig, Germany .,3 Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg , Halle, Germany
| | - Gero Hütter
- 4 CCC Cellex Collection Center GmbH , Dresden, Germany
| | - Bastian Marquass
- 2 Department of Orthopedics, Trauma and Plastic Surgery, University of Leipzig , Leipzig, Germany
| | - Gabriela Aust
- 5 Research Laboratories of the Department of Orthopedics, Trauma and Plastic Surgery, University Hospital of Leipzig AöR , Leipzig, Germany
| | - Ronny M Schulz
- 1 Centre for Biotechnology and Biomedicine, University of Leipzig , Leipzig, Germany .,2 Department of Orthopedics, Trauma and Plastic Surgery, University of Leipzig , Leipzig, Germany
| |
Collapse
|
31
|
Tjin G, Flores-Figueroa E, Duarte D, Straszkowski L, Scott M, Khorshed RA, Purton LE, Lo Celso C. Imaging methods used to study mouse and human HSC niches: Current and emerging technologies. Bone 2019; 119:19-35. [PMID: 29704697 DOI: 10.1016/j.bone.2018.04.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/18/2022]
Abstract
Bone marrow contains numerous different cell types arising from hematopoietic stem cells (HSCs) and non-hematopoietic mesenchymal/skeletal stem cells, in addition to other cell types such as endothelial cells- these non-hematopoietic cells are commonly referred to as stromal cells or microenvironment cells. HSC function is intimately linked to complex signals integrated by their niches, formed by combinations of hematopoietic and stromal cells. Studies of hematopoietic cells have been significantly advanced by flow cytometry methods, enabling the quantitation of each cell type in normal and perturbed situations, in addition to the isolation of these cells for molecular and functional studies. Less is known, however, about the specific niches for distinct developing hematopoietic lineages, or the changes occurring in the niche size and function in these distinct anatomical sites in the bone marrow under stress situations and ageing. Significant advances in imaging technology during the last decade have permitted studies of HSC niches in mice. Additional imaging technologies are emerging that will facilitate the study of human HSC niches in trephine BM biopsies. Here we provide an overview of imaging technologies used to study HSC niches, in addition to highlighting emerging technology that will help us to more precisely identify and characterize HSC niches in normal and diseased states.
Collapse
Affiliation(s)
- Gavin Tjin
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Eugenia Flores-Figueroa
- Oncology Research Unit, Oncology Hospital, National Medical Center Century XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Delfim Duarte
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, London, UK; The Sir Francis Crick Institute, London, UK
| | - Lenny Straszkowski
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Mark Scott
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, London, UK; Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Reema A Khorshed
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, London, UK
| | - Louise E Purton
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St Vincent's Hospital, Fitzroy, Victoria, Australia.
| | - Cristina Lo Celso
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, London, UK; The Sir Francis Crick Institute, London, UK.
| |
Collapse
|
32
|
Prominence of nestin-expressing Schwann cells in bone marrow of patients with myelodysplastic syndromes with severe fibrosis. Int J Hematol 2019; 109:309-318. [PMID: 30632058 DOI: 10.1007/s12185-018-02576-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/05/2018] [Accepted: 12/10/2018] [Indexed: 10/27/2022]
Abstract
Nestin-expressing stromal cells (NESCs) and Schwann cells in the bone marrow (BM) play crucial roles as a niche for normal hematopoietic stem cells in mice. It has been reported that both types of cells are decreased in myeloproliferative neoplasms in patients and also in a mouse model, whereas an increase in NESCs was reported in acute myeloid leukemia. It is thus of interest whether and how these BM stromal cells are structured in myelodysplastic syndromes (MDS). Here, we focused on NESCs and glial fibrillary acidic protein (GFAP)-expressing cells in the BM of MDS patients. We found a marked increase of NESCs in MDS with fibrosis (MDS-F) at a high frequency (9/19; 47.4%), but not in MDS without fibrosis (0/26; 0%). Intriguingly, in eight of the nine (88.9%) MDS-F cases with elevated NESCs, a majority of NESCs also expressed GFAP, with an additional increase in GFAP single-positive cells. Furthermore, in seven of them, we found a prominent structure characterized by neurofilament heavy chain staining surrounded by NESCs with GFAP expression. This structure may represent peripheral nerve axons surrounded by Schwann cells, and could be relevant to the pathophysiology of MDS-F.
Collapse
|
33
|
Roversi FM, Cury NM, Lopes MR, Ferro KP, Machado-Neto JA, Alvarez MC, Dos Santos GP, Giardini Rosa R, Longhini AL, Duarte ADSS, Pericole FV, Favaro P, Yunes JA, Saad STO. Up-regulation of SPINT2/HAI-2 by Azacytidine in bone marrow mesenchymal stromal cells affects leukemic stem cell survival and adhesion. J Cell Mol Med 2018; 23:1562-1571. [PMID: 30484958 PMCID: PMC6349149 DOI: 10.1111/jcmm.14066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 10/30/2018] [Accepted: 11/08/2018] [Indexed: 11/29/2022] Open
Abstract
The role of tumour microenvironment in neoplasm initiation and malignant evolution has been increasingly recognized. However, the bone marrow mesenchymal stromal cell (BMMSC) contribution to disease progression remains poorly explored. We previously reported that the expression of serine protease inhibitor kunitz‐type2 (SPINT2/HAI‐2), an inhibitor of hepatocyte growth factor (HGF) activation, is significantly lower in BMMSC from myelodysplastic syndromes (MDS) patients compared to healthy donors (HD). Thus, to investigate whether this loss of expression was due to SPINT2/HAI‐2 methylation, BMMSC from MDS and de novo acute myeloid leukaemia (de novo AML) patients were treated with 5‐Azacitidine (Aza), a DNA methyltransferase inhibitor. In MDS‐ and de novo AML‐BMMSC, Aza treatment resulted in a pronounced SPINT2/HAI‐2 levels up‐regulation. Moreover, Aza treatment of HD‐BMMSC did not improve SPINT2/HAI‐2 levels. To understand the role of SPINT2/HAI‐2 down‐regulation in BMMSC physiology, SPINT2/HAI‐2 expression was inhibited by lentivirus. SPINT2 underexpression resulted in an increased production of HGF by HS‐5 stromal cells and improved survival of CD34+ de novo AML cells. We also observed an increased adhesion of de novo AML hematopoietic cells to SPINT2/HAI‐2 silenced cells. Interestingly, BMMSC isolated from MDS and de novo AML patients had increased expression of the integrins CD49b, CD49d, and CD49e. Thus, SPINT2/HAI‐2 may contribute to functional and morphological abnormalities of the microenvironment niche and to stem/progenitor cancer cell progression. Hence, down‐regulation in SPINT2/HAI‐2 gene expression, due to methylation in MDS‐BMMSC and de novo AML‐BMMSC, provides novel insights into the pathogenic role of the leukemic bone marrow microenvironment.
Collapse
Affiliation(s)
- Fernanda Marconi Roversi
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-UNICAMP, Campinas, São Paulo, Brazil
| | - Nathalia Moreno Cury
- Centro Infantil de Investigações Hematológicas Dr. Domingos A. Boldrini, Campinas, São Paulo, Brazil
| | - Matheus Rodrigues Lopes
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-UNICAMP, Campinas, São Paulo, Brazil
| | - Karla Priscila Ferro
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-UNICAMP, Campinas, São Paulo, Brazil
| | - João Agostinho Machado-Neto
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-UNICAMP, Campinas, São Paulo, Brazil
| | - Marisa Claudia Alvarez
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-UNICAMP, Campinas, São Paulo, Brazil
| | - Gabriela Pereira Dos Santos
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-UNICAMP, Campinas, São Paulo, Brazil
| | - Renata Giardini Rosa
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-UNICAMP, Campinas, São Paulo, Brazil
| | - Ana Leda Longhini
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-UNICAMP, Campinas, São Paulo, Brazil
| | | | - Fernando Vieira Pericole
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-UNICAMP, Campinas, São Paulo, Brazil
| | - Patricia Favaro
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-UNICAMP, Campinas, São Paulo, Brazil
| | - José Andres Yunes
- Centro Infantil de Investigações Hematológicas Dr. Domingos A. Boldrini, Campinas, São Paulo, Brazil
| | - Sara Teresinha Olalla Saad
- Hematology and Transfusion Medicine Center-University of Campinas/Hemocentro-UNICAMP, Campinas, São Paulo, Brazil
| |
Collapse
|
34
|
Activation of NF-κB driven inflammatory programs in mesenchymal elements attenuates hematopoiesis in low-risk myelodysplastic syndromes. Leukemia 2018; 33:536-541. [PMID: 30315231 PMCID: PMC6365382 DOI: 10.1038/s41375-018-0267-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 07/23/2018] [Accepted: 08/28/2018] [Indexed: 01/19/2023]
Abstract
Activation of NF-κB signaling in mesenchymal cells is common in LR-MDS. Activation of NF-κB in mesenchymal cells leads to transcriptional overexpression of inflammatory factors including negative regulators of hematopoiesis. Activation of NF-κB attenuates HSPC numbers and function ex vivo.
Collapse
|
35
|
Niches for hematopoietic stem cells and immune cell progenitors. Int Immunol 2018; 31:5-11. [DOI: 10.1093/intimm/dxy058] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/29/2018] [Indexed: 01/01/2023] Open
|
36
|
Induction of Expression of CD271 and CD34 in Mesenchymal Stromal Cells Cultured as Spheroids. Stem Cells Int 2018; 2018:7357213. [PMID: 30154865 PMCID: PMC6091361 DOI: 10.1155/2018/7357213] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/18/2018] [Accepted: 07/03/2018] [Indexed: 02/07/2023] Open
Abstract
Cultured mesenchymal stromal cells (MSCs) are cells that can be used for tissue engineering or cell therapies owing to their multipotency and ability to secrete immunomodulatory and trophic molecules. Several studies suggest that MSCs can become pericytes when cocultured with endothelial cells (ECs) but failed to use pericyte markers not already expressed by MSCs. We hypothesized ECs could instruct MSCs to express the molecules CD271 or CD34, which are expressed by pericytes in situ but not by MSCs. CD271 is a marker of especial interest because it is associated with multipotency, a characteristic that wanes in MSCs as they are culture expanded. Consequently, surface expression of CD271 and CD34 was detected in roughly half of the MSCs cocultured with ECs as spheroids in the presence of insulin-like growth factor 1 (IGF-1). Conversely, expression of CD271 and CD34 was detected in a similar proportion of MSCs cultured under these conditions without ECs, and expression of these markers was low or absent when no IGF-1 was added. These findings indicate that specific culture conditions including IGF-1 can endow cultured MSCs with expression of CD271 and CD34, which may enhance the multipotency of these cells when they are used for therapeutic purposes.
Collapse
|
37
|
Petters O, Schmidt C, Thuemmler C, Peinemann F, Zscharnack M, Somerson JS, Schulz RM. Point-of-care treatment of focal cartilage defects with selected chondrogenic mesenchymal stromal cells-An in vitro proof-of-concept study. J Tissue Eng Regen Med 2018; 12:1717-1727. [PMID: 29766671 DOI: 10.1002/term.2699] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 08/16/2017] [Accepted: 05/03/2018] [Indexed: 12/11/2022]
Abstract
Due to the poor self-healing capacities of cartilage, innovative approaches are a major clinical need. The use of in vitro expanded mesenchymal stromal cells (MSCs) in a 2-stage approach is accompanied by cost-, time-, and personnel-intensive good manufacturing practice production. A 1-stage intraoperative procedure could overcome these drawbacks. The aim was to prove the feasibility of a point-of-care concept for the treatment of cartilage lesions using defined MSC subpopulations in a collagen hydrogel without prior MSC monolayer expansion. We tested 4 single marker candidates (MSCA-1, W4A5, CD146, CD271) for their effectiveness of separating colony-forming units of ovine MSCs via magnetic cell separation. The most promising surface marker with regard to the highest enrichment of colony-forming cells was subsequently used to isolate a MSC subpopulation for the direct generation of a cartilage graft composed of a collagen type I hydrogel without the propagation of MSCs in monolayer. We observed that separation with CD271 sustained the highest enrichment of colony-forming units. We then demonstrated the feasibility of generating a cartilage graft with an unsorted bone marrow mononuclear cell fraction and with a characterized CD271 positive MSC subpopulation without the need for a prior cell expansion. A reduced volume of 6.25% of the CD271 positive MSCs was needed to achieve the same results regarding chondrogenesis compared with the unseparated bone marrow mononuclear cell fraction, drastically reducing the number of nonrelevant cells. This study provides a proof-of-concept and reflects the potential of an intraoperative procedure for direct seeding of cartilage grafts with selected CD271 positive cells from bone marrow.
Collapse
Affiliation(s)
- Oliver Petters
- Centre for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany.,Clinic of Orthopedics, Traumatology and Plastic Surgery, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Christian Schmidt
- Centre for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany.,Clinic of Orthopedics, Traumatology and Plastic Surgery, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Christian Thuemmler
- Centre for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany.,Clinic of Orthopedics, Traumatology and Plastic Surgery, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Frank Peinemann
- Centre for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany.,Clinic of Orthopedics, Traumatology and Plastic Surgery, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Matthias Zscharnack
- Centre for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany
| | - Jeremy S Somerson
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, USA
| | - Ronny M Schulz
- Centre for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany.,Clinic of Orthopedics, Traumatology and Plastic Surgery, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
38
|
Goulard M, Dosquet C, Bonnet D. Role of the microenvironment in myeloid malignancies. Cell Mol Life Sci 2018; 75:1377-1391. [PMID: 29222645 PMCID: PMC5852194 DOI: 10.1007/s00018-017-2725-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/01/2017] [Accepted: 12/05/2017] [Indexed: 12/28/2022]
Abstract
The bone marrow microenvironment (BMM) regulates the fate of hematopoietic stem cells (HSCs) in homeostatic and pathologic conditions. In myeloid malignancies, new insights into the role of the BMM and its cellular and molecular actors in the progression of the diseases have started to emerge. In this review, we will focus on describing the major players of the HSC niche and the role of the altered niche function in myeloid malignancies, more specifically focusing on the mesenchymal stroma cell compartment.
Collapse
Affiliation(s)
- Marie Goulard
- INSERM, UMRS1131-Paris Diderot University, Saint Louis Hospital, Paris, France
| | - Christine Dosquet
- INSERM, UMRS1131-Paris Diderot University, Saint Louis Hospital, Paris, France
- Cell Biology Department, APHP, Saint Louis Hospital, Paris, France
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1, Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
39
|
Jakubikova J, Cholujova D, Hideshima T, Gronesova P, Soltysova A, Harada T, Joo J, Kong SY, Szalat RE, Richardson PG, Munshi NC, Dorfman DM, Anderson KC. A novel 3D mesenchymal stem cell model of the multiple myeloma bone marrow niche: biologic and clinical applications. Oncotarget 2018; 7:77326-77341. [PMID: 27764795 PMCID: PMC5357212 DOI: 10.18632/oncotarget.12643] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/28/2016] [Indexed: 12/19/2022] Open
Abstract
Specific niches within the tumor bone marrow (BM) microenvironment afford a sanctuary for multiple myeloma (MM) clones due to stromal cell-tumor cell interactions, which confer survival advantage and drug resistance. Defining the sequelae of tumor cell interactions within the MM niches on an individualized basis may provide the rationale for personalized therapies. To mimic the MM niche, we here describe a new 3D co-culture ex-vivo model in which primary MM patient BM cells are co-cultured with mesenchymal stem cells (MSC) in a hydrogel 3D system. In the 3D model, MSC with conserved phenotype (CD73+CD90+CD105+) formed compact clusters with active fibrous connections, and retained lineage differentiation capacity. Extracellular matrix molecules, integrins, and niche related molecules including N-cadherin and CXCL12 are expressed in 3D MSC model. Furthermore, activation of osteogenesis (MMP13, SPP1, ADAMTS4, and MGP genes) and osteoblastogenic differentiation was confirmed in 3D MSC model. Co-culture of patient-derived BM mononuclear cells with either autologous or allogeneic MSC in 3D model increased proliferation of MM cells, CXCR4 expression, and SP cells. We carried out immune profiling to show that distribution of immune cell subsets was similar in 3D and 2D MSC model systems. Importantly, resistance to novel agents (IMiDs, bortezomib, carfilzomib) and conventional agents (doxorubicin, dexamethasone, melphalan) was observed in 3D MSC system, reflective of clinical resistance. This 3D MSC model may therefore allow for studies of MM pathogenesis and drug resistance within the BM niche. Importantly, ongoing prospective trials are evaluating its utility to inform personalized targeted and immune therapy in MM.
Collapse
Affiliation(s)
- Jana Jakubikova
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA.,Cancer Research Institute, Biomedical Research Center SAS, Bratislava, Slovak Republic
| | - Danka Cholujova
- Cancer Research Institute, Biomedical Research Center SAS, Bratislava, Slovak Republic
| | - Teru Hideshima
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Paulina Gronesova
- Cancer Research Institute, Biomedical Research Center SAS, Bratislava, Slovak Republic
| | - Andrea Soltysova
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Takeshi Harada
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jungnam Joo
- Biometric Research Branch, Division of Cancer Epidemiology and Prevention, Research Institute & Hospital, National Cancer Center, Goyang-si Gyeonggi-do, South Korea
| | - Sun-Young Kong
- Department of Laboratory Medicine and Translational Epidemiology Branch, Research Institute & Hospital, National Cancer Center, Goyang-si Gyeonggi-do, South Korea
| | - Raphael E Szalat
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Paul G Richardson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Nikhil C Munshi
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - David M Dorfman
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kenneth C Anderson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Coexistence of aberrant hematopoietic and stromal elements in myelodysplastic syndromes. Blood Cells Mol Dis 2017; 66:37-46. [PMID: 28822917 DOI: 10.1016/j.bcmd.2017.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/03/2017] [Accepted: 08/07/2017] [Indexed: 11/23/2022]
Abstract
Myelodysplastic syndromes (MDS) are a group of clonal hematopoietic disorders related to hematopoietic stem and progenitor cell dysfunction. Several studies have shown the role of the bone marrow microenvironment in regulating hematopoietic stem, and progenitor function and their individual abnormalities have been associated with disease pathogenesis. In this study, we simultaneously evaluated hematopoietic stem cells (HSC), hematopoietic stem progenitor cells (HSPCs) and different stromal elements in a cohort of patients with MDS-refractory cytopenia with multilineage dysplasia (RCMD). Karyotyping of these patients revealed variable chromosomal abnormalities in 73.33% of patients. Long-term HSC and lineage-negative CD34+CD38- cells were reduced while among the HPCs, there was an expansion of common myeloid progenitor and loss of granulocyte-monocyte progenitors. Interestingly, loss of HSCs was accompanied by aberrant frequencies of endothelial (ECs) (CD31+CD45-CD71-) and mesenchymal stem cells (MSCs) (CD31-CD45-71-) and its subsets associated with HSC niche. We further demonstrate down-regulation of HSC maintenance genes such as Cxcl12, VEGF in mesenchymal cells and a parallel upregulation in endothelial cells. Altogether we report for the first time quantitative and qualitative de novo changes in hematopoietic stem and its associated niche in a cohort of MDS-RCMD patients. These findings further reinforce the role of different components of the bone marrow microenvironment in MDS pathogenesis and emphasize the need for comprehensive simultaneous evaluation of all niche elements in such studies.
Collapse
|
41
|
The microenvironment in human myeloid malignancies: emerging concepts and therapeutic implications. Blood 2017; 129:1617-1626. [PMID: 28159735 DOI: 10.1182/blood-2016-11-696070] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 01/23/2017] [Indexed: 12/13/2022] Open
Abstract
Similar to their healthy counterpart, malignant hematopoietic stem cells in myeloid malignancies, such as myeloproliferative neoplasms, myelodysplastic syndromes, and acute myeloid leukemia, reside in a highly complex and dynamic cellular microenvironment in the bone marrow. This environment provides key regulatory signals for and tightly controls cardinal features of hematopoietic stem cells (HSCs), including self-renewal, quiescence, differentiation, and migration. These features are essential to maintaining cellular homeostasis and blood regeneration throughout life. A large number of studies have extensively addressed the composition of the bone marrow niche in mouse models, as well as the cellular and molecular communication modalities at play under both normal and pathogenic situations. Although instrumental to interrogating the complex composition of the HSC niche and dissecting the niche remodeling processes that appear to actively contribute to leukemogenesis, these models may not fully recapitulate the human system due to immunophenotypic, architectural, and functional inter-species variability. This review summarizes several aspects related to the human hematopoietic niche: (1) its anatomical structure, composition, and function in normal hematopoiesis; (2) its alteration and functional relevance in the context of chronic and acute myeloid malignancies; (3) age-related niche changes and their suspected impact on hematopoiesis; (4) ongoing efforts to develop new models to study niche-leukemic cell interaction in human myeloid malignancies; and finally, (5) how the knowledge gained into leukemic stem cell (LSC) niche dependencies might be exploited to devise novel therapeutic strategies that aim at disrupting essential niche-LSC interactions or improve the regenerative ability of the disease-associated hematopoietic niche.
Collapse
|
42
|
Sá da Bandeira D, Casamitjana J, Crisan M. Pericytes, integral components of adult hematopoietic stem cell niches. Pharmacol Ther 2016; 171:104-113. [PMID: 27908803 DOI: 10.1016/j.pharmthera.2016.11.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The interest in perivascular cells as a niche for adult hematopoietic stem cells (HSCs) is significantly growing. In the adult bone marrow (BM), perivascular cells and HSCs cohabit. Among perivascular cells, pericytes are precursors of mesenchymal stem/stromal cells (MSCs) that are capable of differentiating into osteoblasts, adipocytes and chondrocytes. In situ, pericytes are recognised by their localisation to the abluminal side of the blood vessel wall and closely associated with endothelial cells, in combination with the expression of markers such as CD146, neural glial 2 (NG2), platelet derived growth factor receptor β (PDGFRβ), α-smooth muscle actin (α-SMA), nestin (Nes) and/or leptin receptor (LepR). However, not all pericytes share a common phenotype: different immunophenotypes can be associated with distinct mesenchymal features, including hematopoietic support. In adult BM, arteriolar and sinusoidal pericytes control HSC behaviour, maintenance, quiescence and trafficking through paracrine effects. Different groups identified and characterized hematopoietic supportive pericyte subpopulations using various markers and mouse models. In this review, we summarize recent work performed by others to understand the role of the perivascular niche in the biology of HSCs in adults, as well as their importance in the development of therapies.
Collapse
Affiliation(s)
- D Sá da Bandeira
- BHF Centre for Cardiovascular Science, MRC Scottish Centre for Regenerative Medicine, The Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - J Casamitjana
- BHF Centre for Cardiovascular Science, MRC Scottish Centre for Regenerative Medicine, The Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - M Crisan
- BHF Centre for Cardiovascular Science, MRC Scottish Centre for Regenerative Medicine, The Edinburgh Medical School, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
43
|
Gars E, Yousry SM, Babu D, Kurzer JH, George TI, Gratzinger D. A replicable CD271+ mesenchymal stromal cell density score: bringing the dysfunctional myelodysplastic syndrome niche to the diagnostic laboratory. Leuk Lymphoma 2016; 58:1730-1732. [DOI: 10.1080/10428194.2016.1251590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Eric Gars
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sherif M. Yousry
- Department of Clinical Pathology, Faculty of Medicine, Kasr El-Ainy Hospital, Cairo University, Cairo, Egypt
| | - Daniel Babu
- Department of Pathology, University of New Mexico, University of New Mexico, Albuquerque, NM, USA
| | - Jason H. Kurzer
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tracy I. George
- Department of Pathology, University of New Mexico, University of New Mexico, Albuquerque, NM, USA
| | - Dita Gratzinger
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
44
|
Zambetti N, Ping Z, Chen S, Kenswil K, Mylona M, Sanders M, Hoogenboezem R, Bindels E, Adisty M, Van Strien P, van der Leije C, Westers T, Cremers E, Milanese C, Mastroberardino P, van Leeuwen J, van der Eerden B, Touw I, Kuijpers T, Kanaar R, van de Loosdrecht A, Vogl T, Raaijmakers M. Mesenchymal Inflammation Drives Genotoxic Stress in Hematopoietic Stem Cells and Predicts Disease Evolution in Human Pre-leukemia. Cell Stem Cell 2016; 19:613-627. [DOI: 10.1016/j.stem.2016.08.021] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 07/06/2016] [Accepted: 08/22/2016] [Indexed: 12/22/2022]
|
45
|
da Silva Meirelles L, de Deus Wagatsuma VM, Malta TM, Bonini Palma PV, Araújo AG, Panepucci RA, Silva WA, Kashima S, Covas DT. The gene expression profile of non-cultured, highly purified human adipose tissue pericytes: Transcriptomic evidence that pericytes are stem cells in human adipose tissue. Exp Cell Res 2016; 349:239-254. [PMID: 27789253 DOI: 10.1016/j.yexcr.2016.10.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 10/20/2016] [Accepted: 10/22/2016] [Indexed: 12/15/2022]
Abstract
Pericytes (PCs) are a subset of perivascular cells that can give rise to mesenchymal stromal cells (MSCs) when culture-expanded, and are postulated to give rise to MSC-like cells during tissue repair in vivo. PCs have been suggested to behave as stem cells (SCs) in situ in animal models, although evidence for this role in humans is lacking. Here, we analyzed the transcriptomes of highly purified, non-cultured adipose tissue (AT)-derived PCs (ATPCs) to detect gene expression changes that occur as they acquire MSC characteristics in vitro, and evaluated the hypothesis that human ATPCs exhibit a gene expression profile compatible with an AT SC phenotype. The results showed ATPCs are non-proliferative and express genes characteristic not only of PCs, but also of AT stem/progenitor cells. Additional analyses defined a gene expression signature for ATPCs, and revealed putative novel ATPC markers. Almost all AT stem/progenitor cell genes differentially expressed by ATPCs were not expressed by ATMSCs or culture-expanded ATPCs. Genes expressed by ATMSCs but not by ATPCs were also identified. These findings strengthen the hypothesis that PCs are SCs in vascularized tissues, highlight gene expression changes they undergo as they assume an MSC phenotype, and provide new insights into PC biology.
Collapse
Affiliation(s)
- Lindolfo da Silva Meirelles
- Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo, Rua Tenente Catão Roxo 2501, 14051-140 Ribeirão Preto, SP, Brazil; Laboratory for Stem Cells and Tissue Engineering, PPGBioSaúde, Lutheran University of Brazil, Av. Farroupilha 8001, 92425-900 Canoas, RS, Brazil.
| | - Virgínia Mara de Deus Wagatsuma
- Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo, Rua Tenente Catão Roxo 2501, 14051-140 Ribeirão Preto, SP, Brazil
| | - Tathiane Maistro Malta
- Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo, Rua Tenente Catão Roxo 2501, 14051-140 Ribeirão Preto, SP, Brazil
| | - Patrícia Viana Bonini Palma
- Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo, Rua Tenente Catão Roxo 2501, 14051-140 Ribeirão Preto, SP, Brazil
| | - Amélia Goes Araújo
- Laboratory of Large-Scale Functional Biology (LLSFBio), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo, Rua Tenente Catão Roxo 2501, 14051-140 Ribeirão Preto, SP, Brazil
| | - Rodrigo Alexandre Panepucci
- Laboratory of Large-Scale Functional Biology (LLSFBio), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo, Rua Tenente Catão Roxo 2501, 14051-140 Ribeirão Preto, SP, Brazil
| | - Wilson Araújo Silva
- Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo, Rua Tenente Catão Roxo 2501, 14051-140 Ribeirão Preto, SP, Brazil; Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, 14049-900 Ribeirão Preto, SP, Brazil
| | - Simone Kashima
- Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo, Rua Tenente Catão Roxo 2501, 14051-140 Ribeirão Preto, SP, Brazil
| | - Dimas Tadeu Covas
- Center for Cell-Based Therapy (CEPID/FAPESP), Regional Center for Hemotherapy of Ribeirão Preto, University of São Paulo, Rua Tenente Catão Roxo 2501, 14051-140 Ribeirão Preto, SP, Brazil; Department of Clinical Medicine, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, 14049-900 Ribeirão Preto, SP, Brazil
| |
Collapse
|
46
|
Swords RT, Greenberg PL, Wei AH, Durrant S, Advani AS, Hertzberg MS, Jonas BA, Lewis ID, Rivera G, Gratzinger D, Fan AC, Felsher DW, Cortes JE, Watts JM, Yarranton GT, Walling JM, Lancet JE. KB004, a first in class monoclonal antibody targeting the receptor tyrosine kinase EphA3, in patients with advanced hematologic malignancies: Results from a phase 1 study. Leuk Res 2016; 50:123-131. [PMID: 27736729 DOI: 10.1016/j.leukres.2016.09.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/08/2016] [Accepted: 09/10/2016] [Indexed: 01/01/2023]
Abstract
EphA3 is an Ephrin receptor tyrosine kinase that is overexpressed in most hematologic malignancies. We performed a first-in-human multicenter phase I study of the anti-EphA3 monoclonal antibody KB004 in refractory hematologic malignancies in order to determine safety and tolerability, along with the secondary objectives of pharmacokinetics (PK) and pharmacodynamics (PD) assessments, as well as preliminary assessment of efficacy. Patients were enrolled on a dose escalation phase (DEP) initially, followed by a cohort expansion phase (CEP). KB004 was administered by intravenous infusion on days 1, 8, and 15 of each 21-day cycle in escalating doses. A total of 50 patients (AML 39, MDS/MPN 3, MDS 4, DLBCL 1, MF 3) received KB004 in the DEP; an additional 14 patients were treated on the CEP (AML 8, MDS 6). The most common toxicities were transient grade 1 and grade 2 infusion reactions (IRs) in 79% of patients. IRs were dose limiting above 250mg. Sustained exposure exceeding the predicted effective concentration (1ug/mL) and covering the 7-day interval between doses was achieved above 190mg. Responses were observed in patients with AML, MF, MDS/MPN and MDS. In this study, KB004 was well tolerated and clinically active when given as a weekly infusion.
Collapse
Affiliation(s)
- Ronan T Swords
- Leukemia Program, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, FL, United States.
| | | | - Andrew H Wei
- The Alfred Hospital and Monash University, Melbourne, Australia
| | - Simon Durrant
- The Royal Brisbane and Women's Hospital, Brisbane, Australia
| | | | | | - Brian A Jonas
- Department of Internal Medicine, Division of Hematology and Oncology, University of California Davis School of Medicine, UC Davis Comprehensive Cancer Center, United States
| | - Ian D Lewis
- The Royal Adelaide Hospital, Adelaide, Australia
| | | | | | - Alice C Fan
- Stanford Cancer Institute, Stanford, CA, United States
| | | | - Jorge E Cortes
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Justin M Watts
- Leukemia Program, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, FL, United States
| | - Geoff T Yarranton
- KaloBios Pharmaceuticals, Inc., South San Francisco, CA, United States
| | - Jackie M Walling
- KaloBios Pharmaceuticals, Inc., South San Francisco, CA, United States
| | - Jeffrey E Lancet
- H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| |
Collapse
|
47
|
Pleyer L, Valent P, Greil R. Mesenchymal Stem and Progenitor Cells in Normal and Dysplastic Hematopoiesis-Masters of Survival and Clonality? Int J Mol Sci 2016; 17:ijms17071009. [PMID: 27355944 PMCID: PMC4964385 DOI: 10.3390/ijms17071009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/20/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are malignant hematopoietic stem cell disorders that have the capacity to progress to acute myeloid leukemia (AML). Accumulating evidence suggests that the altered bone marrow (BM) microenvironment in general, and in particular the components of the stem cell niche, including mesenchymal stem cells (MSCs) and their progeny, play a pivotal role in the evolution and propagation of MDS. We here present an overview of the role of MSCs in the pathogenesis of MDS, with emphasis on cellular interactions in the BM microenvironment and related stem cell niche concepts. MSCs have potent immunomodulatory capacities and communicate with diverse immune cells, but also interact with various other cellular components of the microenvironment as well as with normal and leukemic stem and progenitor cells. Moreover, compared to normal MSCs, MSCs in MDS and AML often exhibit altered gene expression profiles, an aberrant phenotype, and abnormal functional properties. These alterations supposedly contribute to the “reprogramming” of the stem cell niche into a disease-permissive microenvironment where an altered immune system, abnormal stem cell niche interactions, and an impaired growth control lead to disease progression. The current article also reviews molecular targets that play a role in such cellular interactions and possibilities to interfere with abnormal stem cell niche interactions by using specific targeted drugs.
Collapse
Affiliation(s)
- Lisa Pleyer
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology & Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Richard Greil
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| |
Collapse
|
48
|
Massive parallel RNA sequencing of highly purified mesenchymal elements in low-risk MDS reveals tissue-context-dependent activation of inflammatory programs. Leukemia 2016; 30:1938-42. [PMID: 27109510 PMCID: PMC5240018 DOI: 10.1038/leu.2016.91] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
49
|
Ewalt MD, Gratzinger D. Selective quantitation of microvessel density reveals sinusoidal expansion in myelodysplastic syndromes. Leuk Lymphoma 2016; 57:2923-2926. [DOI: 10.3109/10428194.2016.1170829] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
50
|
Flores-Figueroa E, Gratzinger D. Beyond the Niche: Myelodysplastic Syndrome Topobiology in the Laboratory and in the Clinic. Int J Mol Sci 2016; 17:553. [PMID: 27089321 PMCID: PMC4849009 DOI: 10.3390/ijms17040553] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 03/26/2016] [Accepted: 04/07/2016] [Indexed: 12/18/2022] Open
Abstract
We review the murine and human microenvironment and hematopoietic stem cell niche in the context of intact bone marrow architecture in man and mouse, both in normal and in myelodysplastic syndrome marrow. We propose that the complexity of the hematopoietic stem cell niche can usefully be approached in the context of its topobiology, and we provide a model that incorporates in vitro and in vivo models as well as in situ findings from intact human marrow to explain the changes seen in myelodysplastic syndrome patients. We highlight the clinical application of the study of the bone marrow microenvironment and its topobiology in myelodysplastic syndromes.
Collapse
Affiliation(s)
- Eugenia Flores-Figueroa
- Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Avenida Cuauhtémoc 330, Colonia Doctores, c.p. 06720 Mexico City, Mexico.
| | - Dita Gratzinger
- Department of Pathology, Stanford University School of Medicine 300 Pasteur Dr., L235, Stanford, CA 94305, USA.
| |
Collapse
|