1
|
Khattab S, El Sorady M, El-Ghandour A, Visani G, Piccaluga PP. Hematopoietic and leukemic stem cells homeostasis: the role of bone marrow niche. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1027-1055. [PMID: 39351440 PMCID: PMC11438561 DOI: 10.37349/etat.2024.00262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/01/2024] [Indexed: 10/04/2024] Open
Abstract
The bone marrow microenvironment (BMM) has highly specialized anatomical characteristics that provide a sanctuary place for hematopoietic stem cells (HSCs) that allow appropriate proliferation, maintenance, and self-renewal capacity. Several cell types contribute to the constitution and function of the bone marrow niche. Interestingly, uncovering the secrets of BMM and its interaction with HSCs in health paved the road for research aiming at better understanding the concept of leukemic stem cells (LSCs) and their altered niche. In fact, they share many signals that are responsible for interactions between LSCs and the bone marrow niche, due to several biological similarities between LSCs and HSCs. On the other hand, LSCs differ from HSCs in their abnormal activation of important signaling pathways that regulate survival, proliferation, drug resistance, invasion, and spread. Targeting these altered niches can help in better treatment choices for hematological malignancies and bone marrow disorders in general and acute myeloid leukemia (AML) in particular. Moreover, targeting those niches may help in decreasing the emergence of drug resistance and lower the relapse rate. In this article, the authors reviewed the most recent literature on bone marrow niches and their relations with either normal HSCs and AML cells/LSC, by focusing on pathogenetic and therapeutic implications.
Collapse
Affiliation(s)
- Shaimaa Khattab
- Biobank of Research, IRCCS Azienda Ospedaliera-Universitaria di Bologna Policlinico di S. Orsola, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Bologna University School of Medicine, 40138 Bologna, Italy
- Medical Research Institute, Hematology department, Alexandria University, Alexandria 21561, Egypt
| | - Manal El Sorady
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria 5310002, Egypt
| | - Ashraf El-Ghandour
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria 5310002, Egypt
| | - Giuseppe Visani
- Hematology and Stem Cell Transplant Center, Azienda Ospedaliera Marche Nord, 61121 Pesaro, Italy
| | - Pier Paolo Piccaluga
- Biobank of Research, IRCCS Azienda Ospedaliera-Universitaria di Bologna Policlinico di S. Orsola, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Bologna University School of Medicine, 40138 Bologna, Italy
| |
Collapse
|
2
|
Lim SU, Lee DW, Kim JH, Kang YJ, Kim IY, Oh IH. Chemical Coaxing of Mesenchymal Stromal Cells by Drug Repositioning for Nestin Induction. Int J Mol Sci 2024; 25:8006. [PMID: 39125577 PMCID: PMC11311338 DOI: 10.3390/ijms25158006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) display heterogeneity in origin and functional role in tissue homeostasis. Subsets of MSCs derived from the neural crest express nestin and serve as niches in bone marrow, but the possibility of coaxing MSCs into nestin-expresing cells for enhanced supportive activity is unclear. In this study, as an approach to the chemical coaxing of MSC functions, we screened libraries of clinically approved chemicals to identify compounds capable of inducing nestin expression in MSCs. Out of 2000 clinical compounds, we chose vorinostat as a candidate to coax the MSCs into neural crest-like fates. When treated with vorinostat, MSCs exhibited a significant increase in the expression of genes involved in the pluripotency and epithelial-mesenchymal transition (EMT), as well as nestin and CD146, the markers for pericytes. In addition, these nestin-induced MSCs exhibited enhanced differentiation towards neuronal cells with the upregulation of neurogenic markers, including SRY-box transcription factor 2 (Sox2), SRY-box transcription factor 10 (Sox10) and microtubule associated protein 2 (Map2) in addition to nestin. Moreover, the coaxed MSCs exhibited enhanced supporting activity for hematopoietic progenitors without supporting leukemia cells. These results demonstrate the feasibility of the drug repositioning of MSCs to induce neural crest-like properties through the chemical coaxing of cell fates.
Collapse
Affiliation(s)
- Sun-Ung Lim
- Catholic High-Performance Cell Therapy Center & Department of Medical Life Science, College of Medicine, The Catholic University of Korea, 222, Banpo-Daero, Seocho-Gu, Seoul 06591, Republic of Korea; (S.-U.L.); (D.-W.L.); (I.-Y.K.)
| | - Dae-Won Lee
- Catholic High-Performance Cell Therapy Center & Department of Medical Life Science, College of Medicine, The Catholic University of Korea, 222, Banpo-Daero, Seocho-Gu, Seoul 06591, Republic of Korea; (S.-U.L.); (D.-W.L.); (I.-Y.K.)
| | - Jung-Ho Kim
- Regen Innopharm Inc., Seoul 06591, Republic of Korea; (J.-H.K.); (Y.-J.K.)
| | - Young-Ju Kang
- Regen Innopharm Inc., Seoul 06591, Republic of Korea; (J.-H.K.); (Y.-J.K.)
| | - In-Yong Kim
- Catholic High-Performance Cell Therapy Center & Department of Medical Life Science, College of Medicine, The Catholic University of Korea, 222, Banpo-Daero, Seocho-Gu, Seoul 06591, Republic of Korea; (S.-U.L.); (D.-W.L.); (I.-Y.K.)
| | - Il-Hoan Oh
- Catholic High-Performance Cell Therapy Center & Department of Medical Life Science, College of Medicine, The Catholic University of Korea, 222, Banpo-Daero, Seocho-Gu, Seoul 06591, Republic of Korea; (S.-U.L.); (D.-W.L.); (I.-Y.K.)
- Regen Innopharm Inc., Seoul 06591, Republic of Korea; (J.-H.K.); (Y.-J.K.)
| |
Collapse
|
3
|
Azevedo PL, Maradei S, de Sá Bigni R, Santos Ramires Aragao J, Abdelhay E, Binato R. SLPI overexpression in hMSCs could be implicated in the HSC gene expression profile in AML. Sci Rep 2024; 14:15550. [PMID: 38969699 PMCID: PMC11226598 DOI: 10.1038/s41598-024-66400-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024] Open
Abstract
Acute myeloid leukaemia (AML) is a severe haematological neoplasm that originates from the transformation of haematopoietic stem cells (HSCs) into leukaemic stem cells (LSCs). The bone marrow (BM) microenvironment, particularly that of mesenchymal stromal cells (hMSCs), plays a crucial role in the maintenance of HSCs. In this context, we explored whether alterations in the secretome of hMSCs derived from AML patients (hMSC-AML) could impact HSC gene expression. Proteomic analysis revealed that the secretome of coculture assays with hMSC-AMLs and HSC from healthy donor is altered, with increased levels of secretory leukocyte protease inhibitor (SLPI), a protein associated with important processes for maintenance of the haematopoietic niche that has already been described to be altered in several tumours. Increased SLPI expression was also observed in the BM plasma of AML patients. Transcriptome analysis of HSCs cocultured with hMSC-AML in comparison with HSCs cocultured with hMSC-HD revealed altered expression of SLPI target genes associated with the cell cycle, proliferation, and apoptosis. Important changes were identified, such as increased expression levels of CCNA2, CCNE2, CCND2, CD133 and CDK1 and decreased levels of CDKN2A and IGFBP3, among others. Overall, these findings suggest that the altered secretome of coculture assays with hMSC-AMLs and HSC from healthy donor, particularly increased SLPI expression, can contribute to gene expression changes in HSCs, potentially influencing important molecular mechanisms related to AML development and progression.
Collapse
Affiliation(s)
- Pedro L Azevedo
- Stem Cell Laboratory, Lab. de Células-Tronco (LCT) Centro, National Cancer Institute (INCA), Praça da Cruz Vermelha 23, 6° andar, Ala C, Rio de Janeiro, RJ, CEP: 20230-130, Brazil.
| | - Simone Maradei
- Bone Marrow Transplantation Unit, National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Ricardo de Sá Bigni
- Haematology Service, National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | | | - Eliana Abdelhay
- Stem Cell Laboratory, Lab. de Células-Tronco (LCT) Centro, National Cancer Institute (INCA), Praça da Cruz Vermelha 23, 6° andar, Ala C, Rio de Janeiro, RJ, CEP: 20230-130, Brazil
| | - Renata Binato
- Stem Cell Laboratory, Lab. de Células-Tronco (LCT) Centro, National Cancer Institute (INCA), Praça da Cruz Vermelha 23, 6° andar, Ala C, Rio de Janeiro, RJ, CEP: 20230-130, Brazil
| |
Collapse
|
4
|
Panting RG, Kotecha RS, Cheung LC. The critical role of the bone marrow stromal microenvironment for the development of drug screening platforms in leukemia. Exp Hematol 2024; 133:104212. [PMID: 38552942 DOI: 10.1016/j.exphem.2024.104212] [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: 12/08/2023] [Revised: 02/21/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024]
Abstract
Extensive research over the past 50 years has resulted in significant improvements in survival for patients diagnosed with leukemia. Despite this, a subgroup of patients harboring high-risk genetic alterations still suffer from poor outcomes. There is a desperate need for new treatments to improve survival, yet consistent failure exists in the translation of in vitro drug development to clinical application. Preclinical screening conventionally utilizes tumor cell monocultures to assess drug activity; however, emerging research has acknowledged the vital role of the tumor microenvironment in treatment resistance and disease relapse. Current co-culture drug screening methods frequently employ fibroblasts as the designated stromal cell component. Alternative stromal cell types that are known to contribute to chemoresistance are often absent in preclinical evaluations of drug efficacy. This review highlights mechanisms of chemoresistance by a range of different stromal constituents present in the bone marrow microenvironment. Utilizing an array of stromal cell types at the early stages of drug screening may enhance the translation of in vitro drug development to clinical use. Ultimately, we highlight the need to consider the bone marrow microenvironment in drug screening platforms for leukemia to develop superior therapies for the treatment of high-risk patients with poor prognostic outcomes.
Collapse
Affiliation(s)
- Rhiannon G Panting
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, Western Australia, Australia; Curtin Medical School, Curtin University, Perth, Western Australia, Australia
| | - Rishi S Kotecha
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, Western Australia, Australia; Curtin Medical School, Curtin University, Perth, Western Australia, Australia; School of Medicine, University of Western Australia, Perth, Western Australia, Australia; Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, Western Australia, Australia
| | - Laurence C Cheung
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, Western Australia, Australia; Curtin Medical School, Curtin University, Perth, Western Australia, Australia; Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.
| |
Collapse
|
5
|
Petinati NA, Sadovskaya AV, Sats NV, Kapranov NM, Davydova YO, Fastova EA, Magomedova AU, Vasilyeva AN, Aleshina OA, Arapidi GP, Shender VO, Smirnov IP, Pobeguts OV, Lagarkova MA, Drize NI, Parovichnikova EN. Molecular Changes in Immunological Characteristics of Bone Marrow Multipotent Mesenchymal Stromal Cells in Lymphoid Neoplasia. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:883-903. [PMID: 38880649 DOI: 10.1134/s0006297924050092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 06/18/2024]
Abstract
Immune system and bone marrow stromal cells play an important role in maintaining normal hematopoiesis. Lymphoid neoplasia disturbs not only development of immune cells, but other immune response mechanisms as well. Multipotent mesenchymal stromal cells (MSCs) of the bone marrow are involved in immune response regulation through both intercellular interactions and secretion of various cytokines. In hematological malignancies, the bone marrow stromal microenvironment, including MSCs, is altered. Aim of this study was to describe the differences of MSCs' immunological function in the patients with acute lymphoblastic leukemia (ALL) and diffuse large B-cell lymphoma (DLBCL). In ALL, malignant cells arise from the early precursor cells localized in bone marrow, while in DLBCL they arise from more differentiated B-cells. In this study, only the DLBCL patients without bone marrow involvement were included. Growth parameters, surface marker expression, genes of interest expression, and secretion pattern of bone marrow MSCs from the patients with ALL and DLBCL at the onset of the disease and in remission were studied. MSCs from the healthy donors of corresponding ages were used as controls. It has been shown that concentration of MSCs in the bone marrow of the patients with ALL is reduced at the onset of the disease and is restored upon reaching remission; in the patients with DLBCL this parameter does not change. Proliferative capacity of MSCs did not change in the patients with ALL; however, the cells of the DLBCL patients both at the onset and in remission proliferated significantly faster than those from the donors. Expression of the membrane surface markers and expression of the genes important for differentiation, immunological status maintenance, and cytokine secretion differed significantly in the MSCs of the patients from those of the healthy donors and depended on nosology of the disease. Secretomes of the MSCs varied greatly; a number of proteins associated with immune response regulation, differentiation, and maintenance of hematopoietic stem cells were depleted in the secretomes of the cells from the patients. Lymphoid neoplasia leads to dramatic changes in the functional immunological status of MSCs.
Collapse
Affiliation(s)
- Nataliya A Petinati
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia.
| | - Aleksandra V Sadovskaya
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
- Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Natalia V Sats
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Nikolai M Kapranov
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Yulia O Davydova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Ekaterina A Fastova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Aminat U Magomedova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Anastasia N Vasilyeva
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Olga A Aleshina
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Georgiy P Arapidi
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, 141700, Russia
| | - Viktoria O Shender
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Igor P Smirnov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Olga V Pobeguts
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Maria A Lagarkova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Nina I Drize
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Elena N Parovichnikova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| |
Collapse
|
6
|
Miller AB, Rodriguez FH, Langenbucher A, Lin L, Bray C, Duquette S, Zhang Y, Goulet D, Lane AA, Weinstock DM, Hemann MT, Manalis SR. Leukemia circulation kinetics revealed through blood exchange method. Commun Biol 2024; 7:483. [PMID: 38643279 PMCID: PMC11032325 DOI: 10.1038/s42003-024-06181-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 04/10/2024] [Indexed: 04/22/2024] Open
Abstract
Leukemias and their bone marrow microenvironments undergo dynamic changes over the course of disease. However, little is known about the circulation kinetics of leukemia cells, nor the impact of specific factors on the clearance of circulating leukemia cells (CLCs) from the blood. To gain a basic understanding of CLC dynamics over the course of disease progression and therapeutic response, we apply a blood exchange method to mouse models of acute leukemia. We find that CLCs circulate in the blood for 1-2 orders of magnitude longer than solid tumor circulating tumor cells. We further observe that: (i) leukemia presence in the marrow can limit the clearance of CLCs in a model of acute lymphocytic leukemia (ALL), and (ii) CLCs in a model of relapsed acute myeloid leukemia (AML) can clear faster than their untreated counterparts. Our approach can also directly quantify the impact of microenvironmental factors on CLC clearance properties. For example, data from two leukemia models suggest that E-selectin, a vascular adhesion molecule, alters CLC clearance. Our research highlights that clearance rates of CLCs can vary in response to tumor and treatment status and provides a strategy for identifying basic processes and factors that govern the kinetics of circulating cells.
Collapse
Affiliation(s)
- Alex B Miller
- Harvard-MIT Department of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Boston, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Felicia H Rodriguez
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adam Langenbucher
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Computation and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lin Lin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christina Bray
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sarah Duquette
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ye Zhang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dan Goulet
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew A Lane
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - David M Weinstock
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Merck and Co., Rahway, NJ, USA
| | - Michael T Hemann
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Scott R Manalis
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
7
|
Vukotić M, Kapor S, Simon F, Cokic V, Santibanez JF. Mesenchymal stromal cells in myeloid malignancies: Immunotherapeutic opportunities. Heliyon 2024; 10:e25081. [PMID: 38314300 PMCID: PMC10837636 DOI: 10.1016/j.heliyon.2024.e25081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/06/2024] Open
Abstract
Myeloid malignancies are clonal disorders of the progenitor cells or hematopoietic stem cells, including acute myeloid leukemia, myelodysplastic syndromes, myeloproliferative malignancies, and chronic myelomonocytic leukemia. Myeloid neoplastic cells affect the proliferation and differentiation of other hematopoietic lineages in the bone marrow and peripheral blood, leading to severe and life-threatening complications. Mesenchymal stromal cells (MSCs) residing in the bone marrow exert immunosuppressive functions by suppressing innate and adaptive immune systems, thus creating a supportive and tolerant microenvironment for myeloid malignancy progression. This review summarizes the significant features of MSCs in myeloid malignancies, including their role in regulating cell growth, cell death, and antineoplastic resistance, in addition to their immunosuppressive contributions. Understanding the implications of MSCs in myeloid malignancies could pave the path for potential use in immunotherapy.
Collapse
Affiliation(s)
- Milica Vukotić
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Suncica Kapor
- Department of Hematology, Clinical Hospital Center “Dr. Dragisa Misovic-Dedinje,” University of Belgrade, Serbia
| | - Felipe Simon
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Millennium Nucleus of Ion Channel-Associated Diseases, Universidad de Chile, Santiago, Chile
| | - Vladan Cokic
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Juan F. Santibanez
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| |
Collapse
|
8
|
Costa A, Breccia M. How to improve treatment-free remission eligibility in chronic myeloid leukaemia? Br J Haematol 2024; 204:434-448. [PMID: 38148564 DOI: 10.1111/bjh.19269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 12/28/2023]
Abstract
The achievement of treatment-free remission (TFR) has become a significant clinical end-point in the management of patients with chronic myeloid leukaemia (CML), providing an opportunity to discontinue therapy with tyrosine kinase inhibitors (TKIs) while maintaining deep molecular response (DMR). Early studies, such as the French STIM trial, have demonstrated that a portion of patients can maintain DMR after treatment cessation, with rates ranging from 40% to 50%, and most relapses occurring within the first 6 months. Key prognostic factors for successful TFR, including treatment duration, duration of DMR, risk scores, and transcript type, have been identified. Optimal patient selection for TFR remains a challenge, but recent research provides insights into potential strategies to increase TFR eligibility. Evidence suggests that early intervention switching to achieve optimal response, treatment combinations, proactive switch in the case of absence of DMR, dose-optimization and induction-maintenance approach can improve molecular responses and, consequently, enhance TFR eligibility. In this review, we report and discuss all the potential therapeutic strategies that may enhance eligibility for a first attempt at TFR, with a particular emphasis on potential future approaches.
Collapse
Affiliation(s)
- Alessandro Costa
- Hematology Unit, Department of Medical Sciences and Public Health, Businco Hospital, University of Cagliari, Cagliari, Italy
| | - Massimo Breccia
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| |
Collapse
|
9
|
Costa A, Scalzulli E, Carmosino I, Ielo C, Bisegna ML, Martelli M, Breccia M. Pharmacotherapeutic advances for chronic myelogenous leukemia: beyond tyrosine kinase inhibitors. Expert Opin Pharmacother 2024; 25:189-202. [PMID: 38488824 DOI: 10.1080/14656566.2024.2331778] [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: 02/07/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024]
Abstract
INTRODUCTION Despite the notable success of tyrosine kinase inhibitors (TKIs) in treating chronic myeloid leukemia (CML), a subset of patients experiences resistance, or relapse after discontinuation. This challenge is attributed to the Ph+ leukemia stem cells (LSCs) pool not fully involved in the inhibition process due to the current therapeutic approach. AREAS COVERED Current pharmacological advancements in CML therapy focus on targeting LSCs, intervening in self-renewal pathways, and exploiting biological vulnerabilities. Beyond BCR::ABL1 inhibition, innovative approaches include immunotherapy, epigenetic modulation, and interference with microenvironmental mechanisms. EXPERT OPINION Diverse therapeutic strategies beyond TKIs are under investigation. Immunotherapy with interferon-α (IFN-α) shows some biological effects, although further research is needed for optimal application in enhancing discontinuation rates. Other compounds were able to mobilize Ph+ LSCs from the bone marrow niche (DPP-IV inhibitor vildagliptin or PAI-1 inhibitor TM5614) increasing the LSC clearance or target the CD26, a Ph+ specific surface receptor. It is noteworthy that the majority of these alternative strategies still incorporate TKIs. In conclusion, novel therapeutic perspectives are emerging for CML, holding the potential for substantial advancements in disease treatment.
Collapse
Affiliation(s)
- Alessandro Costa
- Hematology Unit, Businco Hospital, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Emilia Scalzulli
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Ida Carmosino
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Claudia Ielo
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Maria Laura Bisegna
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Maurizio Martelli
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| | - Massimo Breccia
- Hematology, Department of Translational and Precision Medicine, Az. Policlinico Umberto I-Sapienza University, Rome, Italy
| |
Collapse
|
10
|
Sun W, Lv J, Guo S, Lv M. Cellular microenvironment: a key for tuning mesenchymal stem cell senescence. Front Cell Dev Biol 2023; 11:1323678. [PMID: 38111850 PMCID: PMC10725964 DOI: 10.3389/fcell.2023.1323678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/17/2023] [Indexed: 12/20/2023] Open
Abstract
Mesenchymal stem cells (MSCs) possess the ability to self-renew and differentiate into multiple cell types, making them highly suitable for use as seed cells in tissue engineering. These can be derived from various sources and have been found to play crucial roles in several physiological processes, such as tissue repair, immune regulation, and intercellular communication. However, the limited capacity for cell proliferation and the secretion of senescence-associated secreted phenotypes (SASPs) pose challenges for the clinical application of MSCs. In this review, we provide a comprehensive summary of the senescence characteristics of MSCs and examine the different features of cellular microenvironments studied thus far. Additionally, we discuss the mechanisms by which cellular microenvironments regulate the senescence process of MSCs, offering insights into preserving their functionality and enhancing their effectiveness.
Collapse
Affiliation(s)
| | | | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mengzhu Lv
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
11
|
Giallongo S, Duminuco A, Dulcamare I, Zuppelli T, La Spina E, Scandura G, Santisi A, Romano A, Di Raimondo F, Tibullo D, Palumbo GA, Giallongo C. Engagement of Mesenchymal Stromal Cells in the Remodeling of the Bone Marrow Microenvironment in Hematological Cancers. Biomolecules 2023; 13:1701. [PMID: 38136573 PMCID: PMC10741414 DOI: 10.3390/biom13121701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are a subset of heterogeneous, non-hematopoietic fibroblast-like cells which play important roles in tissue repair, inflammation, and immune modulation. MSCs residing in the bone marrow microenvironment (BMME) functionally interact with hematopoietic stem progenitor cells regulating hematopoiesis. However, MSCs have also emerged in recent years as key regulators of the tumor microenvironment. Indeed, they are now considered active players in the pathophysiology of hematologic malignancies rather than passive bystanders in the hematopoietic microenvironment. Once a malignant event occurs, the BMME acquires cellular, molecular, and epigenetic abnormalities affecting tumor growth and progression. In this context, MSC behavior is affected by signals coming from cancer cells. Furthermore, it has been shown that stromal cells themselves play a major role in several hematological malignancies' pathogenesis. This bidirectional crosstalk creates a functional tumor niche unit wherein tumor cells acquire a selective advantage over their normal counterparts and are protected from drug treatment. It is therefore of critical importance to unveil the underlying mechanisms which activate a protumor phenotype of MSCs for defining the unmasked vulnerabilities of hematological cancer cells which could be pharmacologically exploited to disrupt tumor/MSC coupling. The present review focuses on the current knowledge about MSC dysfunction mechanisms in the BMME of hematological cancers, sustaining tumor growth, immune escape, and cancer progression.
Collapse
Affiliation(s)
- Sebastiano Giallongo
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (S.G.); (G.A.P.); (C.G.)
| | - Andrea Duminuco
- Division of Hematology, AOU Policlinico, 95123 Catania, Italy; (A.D.); (A.S.)
| | - Ilaria Dulcamare
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy;
| | - Tatiana Zuppelli
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (T.Z.); (E.L.S.)
| | - Enrico La Spina
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (T.Z.); (E.L.S.)
| | - Grazia Scandura
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy; (G.S.); (A.R.); (F.D.R.)
| | - Annalisa Santisi
- Division of Hematology, AOU Policlinico, 95123 Catania, Italy; (A.D.); (A.S.)
| | - Alessandra Romano
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy; (G.S.); (A.R.); (F.D.R.)
| | - Francesco Di Raimondo
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy; (G.S.); (A.R.); (F.D.R.)
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (T.Z.); (E.L.S.)
| | - Giuseppe A. Palumbo
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (S.G.); (G.A.P.); (C.G.)
| | - Cesarina Giallongo
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (S.G.); (G.A.P.); (C.G.)
| |
Collapse
|
12
|
Silva FS, Barros-Lima A, Souza-Barros M, Crespo-Neto JA, Santos VGR, Pereira DS, Alves-Hanna FS, Magalhães-Gama F, Faria JAQA, Costa AG. A dual-role for IL-10: From leukemogenesis to the tumor progression in acute lymphoblastic leukemia. Cytokine 2023; 171:156371. [PMID: 37725872 DOI: 10.1016/j.cyto.2023.156371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer in the world, and accounts for 25% of all childhood cancers among children under 15 years of age. Longitudinal studies have shown that children with ALL are born with a deregulated immune response that, together with postnatal environmental exposures, favor the onset of the disease. In this context, IL-10, a key cytokine in the regulation of the immune response, presents itself as a paradoxical mediator, initially influencing the development of ALL through the regulation of inflammatory processes and later on the progression of malignancy, with the increase of this molecule in the leukemia microenvironment. According to the literature, this cytokine plays a critical role in the natural history of the disease and plays an important role in two different though complex scenarios. Thus, in this review, we explore the dual role of IL-10 in ALL, and describe its biological characteristics, immunological mechanisms and genetics, as well as its impact on the leukemia microenvironment and its clinical implications.
Collapse
Affiliation(s)
- Flavio Souza Silva
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Amanda Barros-Lima
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Mateus Souza-Barros
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Juniel Assis Crespo-Neto
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | | | - Daniele Sá Pereira
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil; Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
| | - Fabíola Silva Alves-Hanna
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Fábio Magalhães-Gama
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Instituto René Rachou - Fundação Oswaldo Cruz (FIOCRUZ) Minas, Belo Horizonte, Brazil
| | - Jerusa Araújo Quintão Arantes Faria
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Allyson Guimarães Costa
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil; Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil; Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil; Escola de Enfermagem de Manaus, UFAM, Manaus, Brazil.
| |
Collapse
|
13
|
Sabbah R, Saadi S, Shahar-Gabay T, Gerassy S, Yehudai-Resheff S, Zuckerman T. Abnormal adipogenic signaling in the bone marrow mesenchymal stem cells contributes to supportive microenvironment for leukemia development. Cell Commun Signal 2023; 21:277. [PMID: 37817179 PMCID: PMC10563260 DOI: 10.1186/s12964-023-01231-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/16/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is an aggressive hematological malignancy, associated with unfavorable patient outcome, primarily due to disease relapse. Mesenchymal stem cells (MSCs) residing in the bone marrow (BM) niche are the source of mesenchyma-derived subpopulations, including adipocytes, and osteocytes, that are critical for normal hematopoiesis. This study aimed to characterize BM-derived adipocyte/osteocyte fractions and their crosstalk with AML cells as a potential mechanism underlying leukemogenesis. METHODS BM cell subpopulations derived from primary AML patients were evaluated using humanized ex-vivo and in-vivo models, established for this study. The models comprised AML blasts, normal hematopoietic stem and progenitor cells and mesenchymal stromal subpopulations. ELISA, FACS analysis, colony forming unit assay, whole exome sequencing and real-time qPCR were employed to assess the differentiation capacity, genetic status, gene expression and function of these cell fractions. To explore communication pathways between AML cells and BM subpopulations, levels of signaling mediators, including cytokines and chemokines, were measured using the ProcartaPlex multiplex immunoassay. RESULTS The study revealed deficiencies in adipogenic/osteogenic differentiation of BM-MSCs derived from AML patients, with adipocytes directly promoting survival and clonogenicity of AML cells in-vitro. In whole exome sequencing of BM-MSC/stromal cells, the AHNAK2 gene, associated with the stimulation of adipocyte differentiation, was found to be mutated and significantly under-expressed, implying its abnormal function in AML. The evaluation of communication pathways between AML cells and BM subpopulations demonstrated pronounced alterations in the crosstalk between these cell fractions. This was reflected by significantly elevated levels of signaling mediators cytokines/chemokines, in AML-induced adipocytes/osteocytes compared to non-induced MSCs, indicating abnormal hematopoiesis. Furthermore, in-vivo experiments using a fully humanized 3D scaffold model, showed that AML-induced adipocytes were the dominant component of the tumor microenvironment, providing preferential support to leukemia cell survival and proliferation. CONCLUSIONS This study has disclosed direct contribution of impaired functional, genetic and molecular properties of AML patient-derived adipocytes to effective protection of AML blasts from apoptosis and to stimulation of their growth in vitro and in vivo, which overall leads to disease propagation and relapse. The detected AHNAK2 gene mutations in AML-MSCs point to their involvement in the mechanism underlying abnormal adipogenesis. Video Abstract.
Collapse
Affiliation(s)
- Rawan Sabbah
- Clinical Research Institute at Rambam, Rambam Health Care Campus, 3109601, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, 3109601, Technion, Haifa, Israel
| | - Sahar Saadi
- Clinical Research Institute at Rambam, Rambam Health Care Campus, 3109601, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, 3109601, Technion, Haifa, Israel
| | - Tal Shahar-Gabay
- Clinical Research Institute at Rambam, Rambam Health Care Campus, 3109601, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, 3109601, Technion, Haifa, Israel
| | - Shiran Gerassy
- Clinical Research Institute at Rambam, Rambam Health Care Campus, 3109601, Haifa, Israel
| | - Shlomit Yehudai-Resheff
- Clinical Research Institute at Rambam, Rambam Health Care Campus, 3109601, Haifa, Israel
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, 8, Ha'Aliya Street, 3109601, Haifa, Israel
| | - Tsila Zuckerman
- Clinical Research Institute at Rambam, Rambam Health Care Campus, 3109601, Haifa, Israel.
- The Ruth and Bruce Rappaport Faculty of Medicine, 3109601, Technion, Haifa, Israel.
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, 8, Ha'Aliya Street, 3109601, Haifa, Israel.
| |
Collapse
|
14
|
Sandhow L, Cai H, Leonard E, Xiao P, Tomaipitinca L, Månsson A, Kondo M, Sun X, Johansson AS, Tryggvason K, Kasper M, Järås M, Qian H. Skin mesenchymal niches maintain and protect AML-initiating stem cells. J Exp Med 2023; 220:e20220953. [PMID: 37516911 PMCID: PMC10373345 DOI: 10.1084/jem.20220953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 05/10/2023] [Accepted: 06/29/2023] [Indexed: 07/31/2023] Open
Abstract
Leukemia cutis or leukemic cell infiltration in skin is one of the common extramedullary manifestations of acute myeloid leukemia (AML) and signifies a poorer prognosis. However, its pathogenesis and maintenance remain understudied. Here, we report massive AML cell infiltration in the skin in a transplantation-induced MLL-AF9 AML mouse model. These AML cells could regenerate AML after transplantation. Prospective niche characterization revealed that skin harbored mesenchymal progenitor cells (MPCs) with a similar phenotype as BM mesenchymal stem cells. These skin MPCs protected AML-initiating stem cells (LSCs) from chemotherapy in vitro partially via mitochondrial transfer. Furthermore, Lama4 deletion in skin MPCs promoted AML LSC proliferation and chemoresistance. Importantly, more chemoresistant AML LSCs appeared to be retained in Lama4-/- mouse skin after cytarabine treatment. Our study reveals the characteristics and previously unrecognized roles of skin mesenchymal niches in maintaining and protecting AML LSCs during chemotherapy, meriting future exploration of their impact on AML relapse.
Collapse
Affiliation(s)
- Lakshmi Sandhow
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Huan Cai
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Elory Leonard
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Pingnan Xiao
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Luana Tomaipitinca
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Alma Månsson
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Makoto Kondo
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Xiaoyan Sun
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Anne-Sofie Johansson
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Karl Tryggvason
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Maria Kasper
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Marcus Järås
- Department of Clinical Genetics, Lund University, Lund, Sweden
| | - Hong Qian
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
15
|
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
|
16
|
Miller AB, Langenbucher A, Rodriguez FH, Lin L, Bray C, Duquette S, Zhang Y, Goulet D, Lane AA, Weinstock DM, Hemann MT, Manalis SR. Leukemia circulation kinetics revealed through blood exchange method. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.03.556043. [PMID: 37732189 PMCID: PMC10508764 DOI: 10.1101/2023.09.03.556043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Leukemias and their bone marrow microenvironment are known to undergo dynamic changes over the course of disease. However, relatively little is known about the circulation kinetics of leukemia cells, nor the impact of specific factors on the clearance of circulating leukemia cells (CLCs) from the blood. To gain a basic understanding of leukemia cell dynamics over the course of disease progression and therapeutic response, we apply a blood exchange method to mouse models of acute leukemia. We find that CLCs circulate in the blood for 1-2 orders of magnitude longer than solid tumor circulating tumor cells. We further observe that: i) leukemia presence in the marrow can limit the clearance of CLCs in a model of acute lymphocytic leukemia (ALL), and ii) CLCs in a model of relapsed acute myeloid leukemia (AML) can clear faster than their untreated counterparts. Our approach can also directly quantify the impact of microenvironmental factors on CLC clearance properties. For example, data from two leukemia models suggest that E-selectin, a vascular adhesion molecule, alters CLC clearance. Our research highlights that clearance rates of CLCs can vary in response to tumor and treatment status and provides a strategy for identifying basic processes and factors that govern the kinetics of circulating cells.
Collapse
Affiliation(s)
- Alex B Miller
- Harvard-MIT Department of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Boston, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adam Langenbucher
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Computation and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Felicia H Rodriguez
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lin Lin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christina Bray
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sarah Duquette
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ye Zhang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dan Goulet
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew A Lane
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - David M Weinstock
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Michael T Hemann
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Scott R Manalis
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
17
|
Yuan N, Wei W, Ji L, Qian J, Jin Z, Liu H, Xu L, Li L, Zhao C, Gao X, He Y, Wang M, Tang L, Fang Y, Wang J. Young donor hematopoietic stem cells revitalize aged or damaged bone marrow niche by transdifferentiating into functional niche cells. Aging Cell 2023; 22:e13889. [PMID: 37226323 PMCID: PMC10410009 DOI: 10.1111/acel.13889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 05/26/2023] Open
Abstract
The bone marrow niche maintains hematopoietic stem cell (HSC) homeostasis and declines in function in the physiologically aging population and in patients with hematological malignancies. A fundamental question is now whether and how HSCs are able to renew or repair their niche. Here, we show that disabling HSCs based on disrupting autophagy accelerated niche aging in mice, whereas transplantation of young, but not aged or impaired, donor HSCs normalized niche cell populations and restored niche factors in host mice carrying an artificially harassed niche and in physiologically aged host mice, as well as in leukemia patients. Mechanistically, HSCs, identified using a donor lineage fluorescence-tracing system, transdifferentiate in an autophagy-dependent manner into functional niche cells in the host that include mesenchymal stromal cells and endothelial cells, previously regarded as "nonhematopoietic" sources. Our findings thus identify young donor HSCs as a primary parental source of the niche, thereby suggesting a clinical solution to revitalizing aged or damaged bone marrow hematopoietic niche.
Collapse
Affiliation(s)
- Na Yuan
- Research Center for Blood Engineering and ManufacturingCyrus Tang Medical Institute, Suzhou Medical College of Soochow UniversitySuzhouChina
- State Key Laboratory of Radiation Medicine and ProtectionNational Research Center for Hematological Diseases, Collaborative Innovation Center of Hematology, Soochow UniversitySuzhouChina
- The Department of OrthopedicsThe Affiliated Ninth Suzhou Hospital of Soochow UniversitySuzhouChina
| | - Wen Wei
- Research Center for Blood Engineering and ManufacturingCyrus Tang Medical Institute, Suzhou Medical College of Soochow UniversitySuzhouChina
- State Key Laboratory of Radiation Medicine and ProtectionNational Research Center for Hematological Diseases, Collaborative Innovation Center of Hematology, Soochow UniversitySuzhouChina
- The Department of OrthopedicsThe Affiliated Ninth Suzhou Hospital of Soochow UniversitySuzhouChina
| | - Li Ji
- Research Center for Blood Engineering and ManufacturingCyrus Tang Medical Institute, Suzhou Medical College of Soochow UniversitySuzhouChina
| | - Jiawei Qian
- Research Center for Blood Engineering and ManufacturingCyrus Tang Medical Institute, Suzhou Medical College of Soochow UniversitySuzhouChina
| | - Zhicong Jin
- Research Center for Blood Engineering and ManufacturingCyrus Tang Medical Institute, Suzhou Medical College of Soochow UniversitySuzhouChina
| | - Hong Liu
- State Key Laboratory of Radiation Medicine and ProtectionNational Research Center for Hematological Diseases, Collaborative Innovation Center of Hematology, Soochow UniversitySuzhouChina
- Institute of Blood and Marrow Transplantation, Jiangsu Institute of HematologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Li Xu
- Research Center for Blood Engineering and ManufacturingCyrus Tang Medical Institute, Suzhou Medical College of Soochow UniversitySuzhouChina
- State Key Laboratory of Radiation Medicine and ProtectionNational Research Center for Hematological Diseases, Collaborative Innovation Center of Hematology, Soochow UniversitySuzhouChina
| | - Lei Li
- Research Center for Blood Engineering and ManufacturingCyrus Tang Medical Institute, Suzhou Medical College of Soochow UniversitySuzhouChina
| | - Chen Zhao
- Research Center for Blood Engineering and ManufacturingCyrus Tang Medical Institute, Suzhou Medical College of Soochow UniversitySuzhouChina
| | - Xueqin Gao
- Research Center for Blood Engineering and ManufacturingCyrus Tang Medical Institute, Suzhou Medical College of Soochow UniversitySuzhouChina
| | - Yulong He
- Research Center for Blood Engineering and ManufacturingCyrus Tang Medical Institute, Suzhou Medical College of Soochow UniversitySuzhouChina
- State Key Laboratory of Radiation Medicine and ProtectionNational Research Center for Hematological Diseases, Collaborative Innovation Center of Hematology, Soochow UniversitySuzhouChina
| | | | | | - Yixuan Fang
- Research Center for Blood Engineering and ManufacturingCyrus Tang Medical Institute, Suzhou Medical College of Soochow UniversitySuzhouChina
- State Key Laboratory of Radiation Medicine and ProtectionNational Research Center for Hematological Diseases, Collaborative Innovation Center of Hematology, Soochow UniversitySuzhouChina
- The Department of OrthopedicsThe Affiliated Ninth Suzhou Hospital of Soochow UniversitySuzhouChina
| | - Jianrong Wang
- Research Center for Blood Engineering and ManufacturingCyrus Tang Medical Institute, Suzhou Medical College of Soochow UniversitySuzhouChina
- State Key Laboratory of Radiation Medicine and ProtectionNational Research Center for Hematological Diseases, Collaborative Innovation Center of Hematology, Soochow UniversitySuzhouChina
- The Department of OrthopedicsThe Affiliated Ninth Suzhou Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
18
|
Tian C, Chen Z, Wang L, Si J, Kang J, Li Y, Zheng Y, Gao Y, Nuermaimaiti R, You MJ, Zheng G. Over expression of ubiquitin-conjugating enzyme E2O in bone marrow mesenchymal stromal cells partially attenuates acute myeloid leukaemia progression. Br J Haematol 2023; 200:476-488. [PMID: 36345807 DOI: 10.1111/bjh.18541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022]
Abstract
Bone marrow mesenchymal stromal cells (BM-MSCs) are implicated in the pathogenesis of acute myeloid leukaemia (AML). However, due to the high heterogeneity of AML the mechanism underlying the cross-talk between MSCs and leukaemia cells is not well understood. We found that mixed-lineage leukaemia-AF9 (MLL-AF9)-induced AML mice-derived MSCs had higher proliferative viability compared to wild-type mice-derived MSCs with ubiquitin-conjugating enzyme E2O (Ube2o) down-regulation. After overexpression of UBE2O in AML-derived MSCs, the growth capacity of MSCs was reduced with nuclear factor kappa B subunit 1 (NF-κB) pathway deactivation. In vitro co-culture assay revealed that UBE2O-overexpression MSCs suppressed the proliferation and promoted apoptosis of AML cells by direct contact. In vivo results revealed that the leukaemia burden was reduced and the overall survival of AML mice was prolonged, with decreased dissemination of leukaemia cells in BM, spleen, liver and peripheral blood. Additionally, subcutaneous tumorigenesis revealed that tumour growth was also suppressed in the UBE2O-overexpression MSCs group. In conclusion, UBE2O was expressed at a low level in MLL-AF9-induced AML mice-derived MSCs. Overexpression of UBE2O in MSCs suppressed their proliferation through NF-κB pathway deactivation, which resulted in AML suppression. Our study provides a theoretical basis for a BM microenvironment-based therapeutic strategy to control disease progression.
Collapse
Affiliation(s)
- Chen Tian
- Department of Haematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Haematology, Hotan District People's Hospital, Hotan, China
| | - Zehui Chen
- Department of Haematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,State Key Laboratory of Experimental Haematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Haematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Lina Wang
- State Key Laboratory of Experimental Haematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Haematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Junqi Si
- Department of Haematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Junnan Kang
- Department of Haematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,State Key Laboratory of Experimental Haematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Haematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yueyang Li
- Department of Haematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,State Key Laboratory of Experimental Haematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Haematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yaxin Zheng
- Department of Haematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yanan Gao
- Department of Haematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | | | - M James You
- Department of Haematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Guoguang Zheng
- State Key Laboratory of Experimental Haematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Haematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
19
|
Sadovskaya AV, Petinati NA, Kapranov NM, Drize NI, Vasil'eva AN, Aleshina OA, Parovichnikova EN. Dynamics of Changes in the Properties of Multipotent Mesenchymal Stromal Cells in Patients with Acute Leukemia. Bull Exp Biol Med 2023; 174:556-563. [PMID: 36894815 DOI: 10.1007/s10517-023-05747-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Indexed: 03/11/2023]
Abstract
In acute leukemia, the stromal microenvironment of the bone marrow that regulates hematopoiesis is modified under the influence of malignant cells. Chemotherapy also adversely affects stromal cells. Multipotent mesenchymal stromal cells (MSC) are involved in the formation of the stromal microenvironment and in the regulation of normal and tumor hematopoietic cells. The properties of MSC from the bone marrow of patients with acute myeloid and lymphoid leukemia were studied at the onset of the disease and after achieving remission. The immunophenotype and the level of gene expression were analyzed in MSC of 34 patients. In MSC from patients with acute leukemia, the expression of CD105 and CD274 was significantly reduced in comparison with MSC from healthy donors. At the onset of the disease, the expression of IL6, JAG1, PPARG, IGF1, and PDGFRA was enhanced, while the expression of IL1B, IL8, SOX9, ANG1, and TGFB was reduced. All these changes affect the course of the disease in patients and can be the targets of therapeutic intervention.
Collapse
Affiliation(s)
- A V Sadovskaya
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - N A Petinati
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - N M Kapranov
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - N I Drize
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A N Vasil'eva
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - O A Aleshina
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E N Parovichnikova
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
20
|
Bouligny IM, Maher KR, Grant S. Mechanisms of myeloid leukemogenesis: Current perspectives and therapeutic objectives. Blood Rev 2023; 57:100996. [PMID: 35989139 PMCID: PMC10693933 DOI: 10.1016/j.blre.2022.100996] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/28/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematopoietic neoplasm which results in clonal proliferation of abnormally differentiated hematopoietic cells. In this review, mechanisms contributing to myeloid leukemogenesis are summarized, highlighting aberrations of epigenetics, transcription factors, signal transduction, cell cycling, and the bone marrow microenvironment. The mechanisms contributing to AML are detailed to spotlight recent findings that convey clinical impact. The applications of current and prospective therapeutic targets are accentuated in addition to reviews of treatment paradigms stratified for each characteristic molecular lesion - with a focus on exploring novel treatment approaches and combinations to improve outcomes in AML.
Collapse
Affiliation(s)
- Ian M Bouligny
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| | - Keri R Maher
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| | - Steven Grant
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
21
|
Chen Y, Li J, Xu L, Găman MA, Zou Z. The genesis and evolution of acute myeloid leukemia stem cells in the microenvironment: From biology to therapeutic targeting. Cell Death Discov 2022; 8:397. [PMID: 36163119 PMCID: PMC9513079 DOI: 10.1038/s41420-022-01193-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/12/2022] [Accepted: 09/16/2022] [Indexed: 11/09/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by cytogenetic and genomic alterations. Up to now, combination chemotherapy remains the standard treatment for leukemia. However, many individuals diagnosed with AML develop chemotherapeutic resistance and relapse. Recently, it has been pointed out that leukemic stem cells (LSCs) are the fundamental cause of drug resistance and AML relapse. LSCs only account for a small subpopulation of all leukemic cells, but possess stem cell properties, including a self-renewal capacity and a multi-directional differentiation potential. LSCs reside in a mostly quiescent state and are insensitive to chemotherapeutic agents. When LSCs reside in a bone marrow microenvironment (BMM) favorable to their survival, they engage into a steady, continuous clonal evolution to better adapt to the action of chemotherapy. Most chemotherapeutic drugs can only eliminate LSC-derived clones, reducing the number of leukemic cells in the BM to a normal range in order to achieve complete remission (CR). LSCs hidden in the BM niche can hardly be targeted or eradicated, leading to drug resistance and AML relapse. Understanding the relationship between LSCs, the BMM, and the generation and evolution laws of LSCs can facilitate the development of effective therapeutic targets and increase the efficiency of LSCs elimination in AML.
Collapse
Affiliation(s)
- Yongfeng Chen
- Department of Basic Medical Sciences, Medical College of Taizhou University, Taizhou, Zhejiang, 318000, China.
| | - Jing Li
- Department of Histology and Embryology, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Linglong Xu
- Department of Hematology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, 318000, China
| | - Mihnea-Alexandru Găman
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 050474, Bucharest, Romania.
- Department of Hematology, Centre of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, Bucharest, Romania.
| | - Zhenyou Zou
- Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou, Guangxi, 545005, China.
| |
Collapse
|
22
|
Tan Z, Kan C, Wong M, Sun M, Liu Y, Yang F, Wang S, Zheng H. Regulation of Malignant Myeloid Leukemia by Mesenchymal Stem Cells. Front Cell Dev Biol 2022; 10:857045. [PMID: 35756991 PMCID: PMC9213747 DOI: 10.3389/fcell.2022.857045] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Bone marrow microenvironment (BMM) has been proven to have benefits for both normal hematopoietic stem cell niche and pathological leukemic stem cell niche. In fact, the pathological leukemia microenvironment reprograms bone marrow niche cells, especially mesenchymal stem cells for leukemia progression, chemoresistance and relapse. The growth and differentiation of MSCs are modulated by leukemia stem cells. Moreover, chromatin abnormality of mesenchymal stem cells is sufficient for leukemia initiation. Here, we summarize the detailed relationship between MSC and leukemia. MSCs can actively and passively regulate the progression of myelogenous leukemia through cell-to-cell contact, cytokine-receptor interaction, and exosome communication. These behaviors benefit LSCs proliferation and survival and inhibit physiological hematopoiesis. Finally, we describe the recent advances in therapy targeting MSC hoping to provide new perspectives and therapeutic strategies for leukemia.
Collapse
Affiliation(s)
- Zhenya Tan
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Chen Kan
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Mandy Wong
- Department of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Minqiong Sun
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Yakun Liu
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Fan Yang
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Siying Wang
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Hong Zheng
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| |
Collapse
|
23
|
Leukemic Stem Cells as a Target for Eliminating Acute Myeloid Leukemia: Gaps in Translational Research. Crit Rev Oncol Hematol 2022; 175:103710. [DOI: 10.1016/j.critrevonc.2022.103710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 04/29/2022] [Accepted: 05/11/2022] [Indexed: 12/26/2022] Open
|
24
|
Ruiz-Aparicio PF, Vernot JP. Bone Marrow Aging and the Leukaemia-Induced Senescence of Mesenchymal Stem/Stromal Cells: Exploring Similarities. J Pers Med 2022; 12:jpm12050716. [PMID: 35629139 PMCID: PMC9147878 DOI: 10.3390/jpm12050716] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 12/17/2022] Open
Abstract
Bone marrow aging is associated with multiple cellular dysfunctions, including perturbed haematopoiesis, the propensity to haematological transformation, and the maintenance of leukaemia. It has been shown that instructive signals from different leukemic cells are delivered to stromal cells to remodel the bone marrow into a supportive leukemic niche. In particular, cellular senescence, a physiological program with both beneficial and deleterious effects on the health of the organisms, may be responsible for the increased incidence of haematological malignancies in the elderly and for the survival of diverse leukemic cells. Here, we will review the connection between BM aging and cellular senescence and the role that these processes play in leukaemia progression. Specifically, we discuss the role of mesenchymal stem cells as a central component of the supportive niche. Due to the specificity of the genetic defects present in leukaemia, one would think that bone marrow alterations would also have particular changes, making it difficult to envisage a shared therapeutic use. We have tried to summarize the coincident features present in BM stromal cells during aging and senescence and in two different leukaemias, acute myeloid leukaemia, with high frequency in the elderly, and B-acute lymphoblastic leukaemia, mainly a childhood disease. We propose that mesenchymal stem cells are similarly affected in these different leukaemias, and that the changes that we observed in terms of cellular function, redox balance, genetics and epigenetics, soluble factor repertoire and stemness are equivalent to those occurring during BM aging and cellular senescence. These coincident features may be used to explore strategies useful to treat various haematological malignancies.
Collapse
Affiliation(s)
- Paola Fernanda Ruiz-Aparicio
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
| | - Jean-Paul Vernot
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
- Instituto de Investigaciones Biomédicas, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia
- Correspondence:
| |
Collapse
|
25
|
Shao X, Chen Y, Wang W, Du W, Zhang X, Cai M, Bing S, Cao J, Xu X, Yang B, He Q, Ying M. Blockade of deubiquitinase YOD1 degrades oncogenic PML/RARα and eradicates acute promyelocytic leukemia cells. Acta Pharm Sin B 2022; 12:1856-1870. [PMID: 35847510 PMCID: PMC9279643 DOI: 10.1016/j.apsb.2021.10.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/06/2021] [Accepted: 09/28/2021] [Indexed: 11/26/2022] Open
Abstract
In most acute promyelocytic leukemia (APL) cells, promyelocytic leukemia (PML) fuses to retinoic acid receptor α (RARα) due to chromosomal translocation, thus generating PML/RARα oncoprotein, which is a relatively stable oncoprotein for degradation in APL. Elucidating the mechanism regulating the stability of PML/RARα may help to degrade PML/RARα and eradicate APL cells. Here, we describe a deubiquitinase (DUB)-involved regulatory mechanism for the maintenance of PML/RARα stability and develop a novel pharmacological approach to degrading PML/RARα by inhibiting DUB. We utilized a DUB siRNA library to identify the ovarian tumor protease (OTU) family member deubiquitinase YOD1 as a critical DUB of PML/RARα. Suppression of YOD1 promoted the degradation of PML/RARα, thus inhibiting APL cells and prolonging the survival time of APL cell-bearing mice. Subsequent phenotypic screening of small molecules allowed us to identify ubiquitin isopeptidase inhibitor I (G5) as the first YOD1 pharmacological inhibitor. As expected, G5 notably degraded PML/RARα protein and eradicated APL, particularly drug-resistant APL cells. Importantly, G5 also showed a strong killing effect on primary patient-derived APL blasts. Overall, our study not only reveals the DUB-involved regulatory mechanism on PML/RARα stability and validates YOD1 as a potential therapeutic target for APL, but also identifies G5 as a YOD1 inhibitor and a promising candidate for APL, particularly drug-resistant APL treatment.
Collapse
|
26
|
Tan L, Liu X, Dou H, Hou Y. Characteristics and regulation of mesenchymal stem cell plasticity by the microenvironment — specific factors involved in the regulation of MSC plasticity. Genes Dis 2022; 9:296-309. [PMID: 35224147 PMCID: PMC8843883 DOI: 10.1016/j.gendis.2020.10.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/05/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs), multipotent stromal cells, have attracted extensive attention in the field of regenerative medicine and cell therapy due to the capacity of self-renewal, multilineage differentiation, and immune regulation. MSCs have different cellular effects in different diseases, and even have markedly different curative effects with different tissue sources, indicating the plasticity of MSCs. The phenotypes, secreted factors, and proliferative, migratory, differentiating, and immunomodulatory effects of MSCs depend on certain mediators present in their microenvironment. Understanding microenvironmental factors and their internal mechanisms in MSC responses may help in subsequent prediction and improvement of clinical benefits. This review highlighted the recent advances in MSC plasticity in the physiological and pathological microenvironment and multiple microenvironmental factors regulating MSC plasticity. It also highlighted some progress in the underlying molecular mechanisms of MSC remodeling in the microenvironment. It might provide references for the improvement in vitro culture of MSCs, clinical application, and in vivo induction.
Collapse
|
27
|
Roux B, Picou F, Debeissat C, Koubi M, Gallay N, Hirsch P, Ravalet N, Béné MC, Maigre M, Hunault M, Mosser J, Etcheverry A, Gyan E, Delhommeau F, Domenech J, Herault O. Aberrant DNA methylation impacts HOX genes expression in bone marrow mesenchymal stromal cells of myelodysplastic syndromes and de novo acute myeloid leukemia. Cancer Gene Ther 2022; 29:1263-1275. [PMID: 35194200 DOI: 10.1038/s41417-022-00441-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/12/2021] [Accepted: 02/08/2022] [Indexed: 11/09/2022]
Abstract
DNA methylation, a major biological process regulating the transcription, contributes to the pathophysiology of hematologic malignancies, and hypomethylating agents are commonly used to treat myelodysplastic syndromes (MDS) and acute myeloid leukemias (AML). In these diseases, bone marrow mesenchymal stromal cells (MSCs) play a key supportive role through the production of various signals and interactions. The DNA methylation status of MSCs, likely to reflect their functionality, might be relevant to understand their contribution to the pathophysiology of these diseases. Consequently, the aim of our study was to analyze the modifications of DNA methylation profiles of MSCs induced by MDS or AML. MSCs from MDS/AML patients were characterized via 5-methylcytosine quantification, gene expression profiles of key regulators of DNA methylation, identification of differentially methylated regions (DMRs) by methylome array, and quantification of DMR-coupled genes expression. MDS and AML-MSCs displayed global hypomethylation and under-expression of DNMT1 and UHRF1. Methylome analysis revealed aberrant methylation profiles in all MDS and in a subgroup of AML-MSCs. This aberrant methylation was preferentially found in the sequence of homeobox genes, especially from the HOX family (HOXA1, HOXA4, HOXA5, HOXA9, HOXA10, HOXA11, HOXB5, HOXC4, and HOXC6), and impacted on their expression. These results highlight modifications of DNA methylation in MDS/AML-MSCs, both at global and focal levels dysregulating the expression of HOX genes well known for their involvement in leukemogenesis. Such DNA methylation in MSCs could be the consequence of the malignant disease or could participate in its development through defective functionality or exosomal transfer of HOX transcription factors from MSCs to hematopoietic cells.
Collapse
Affiliation(s)
- Benjamin Roux
- CNRS EMR 7001 LNOx "Leukemic niche & redox metabolism", Tours, France.,EA 7501 GICC, université de Tours, Tours, France.,CHU de Tours, Service d'Hématologie Biologique, Tours, France
| | - Frédéric Picou
- CNRS EMR 7001 LNOx "Leukemic niche & redox metabolism", Tours, France.,EA 7501 GICC, université de Tours, Tours, France.,CHU de Tours, Service d'Hématologie Biologique, Tours, France
| | - Christelle Debeissat
- CNRS EMR 7001 LNOx "Leukemic niche & redox metabolism", Tours, France.,EA 7501 GICC, université de Tours, Tours, France.,CHU de Tours, Service d'Hématologie Biologique, Tours, France
| | - Myriam Koubi
- CNRS EMR 7001 LNOx "Leukemic niche & redox metabolism", Tours, France.,EA 7501 GICC, université de Tours, Tours, France
| | - Nathalie Gallay
- CNRS EMR 7001 LNOx "Leukemic niche & redox metabolism", Tours, France.,EA 7501 GICC, université de Tours, Tours, France.,CHU de Tours, Service d'Hématologie Biologique, Tours, France
| | - Pierre Hirsch
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France
| | - Noémie Ravalet
- CNRS EMR 7001 LNOx "Leukemic niche & redox metabolism", Tours, France.,EA 7501 GICC, université de Tours, Tours, France.,CHU de Tours, Service d'Hématologie Biologique, Tours, France
| | - Marie C Béné
- CHU de Nantes, Service d'Hématologie Biologique, CRCINA, Nantes, France.,FHU GOAL, Angers, France
| | | | - Mathilde Hunault
- FHU GOAL, Angers, France.,CHU d'Angers, Service d'Hématologie, Angers, France
| | - Jean Mosser
- CHU de Rennes, Service de Génétique Moléculaire et Génomique, Rennes, France.,Cancéropôle Grand Ouest, Nantes, France
| | - Amandine Etcheverry
- CHU de Rennes, Service de Génétique Moléculaire et Génomique, Rennes, France
| | - Emmanuel Gyan
- CNRS EMR 7001 LNOx "Leukemic niche & redox metabolism", Tours, France.,EA 7501 GICC, université de Tours, Tours, France.,CHU de Tours, Service d'Hématologie et Thérapie Cellulaire, Tours, France
| | - François Delhommeau
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Biologique, Paris, France.,CNRS GDR 3697 Micronit "Microenvironment of tumor niches", Tours, France.,OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Jorge Domenech
- CNRS EMR 7001 LNOx "Leukemic niche & redox metabolism", Tours, France.,EA 7501 GICC, université de Tours, Tours, France.,CHU de Tours, Service d'Hématologie Biologique, Tours, France
| | - Olivier Herault
- CNRS EMR 7001 LNOx "Leukemic niche & redox metabolism", Tours, France. .,EA 7501 GICC, université de Tours, Tours, France. .,CHU de Tours, Service d'Hématologie Biologique, Tours, France. .,FHU GOAL, Angers, France. .,Cancéropôle Grand Ouest, Nantes, France. .,CNRS GDR 3697 Micronit "Microenvironment of tumor niches", Tours, France. .,OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France.
| |
Collapse
|
28
|
Mancini SJC, Balabanian K, Corre I, Gavard J, Lazennec G, Le Bousse-Kerdilès MC, Louache F, Maguer-Satta V, Mazure NM, Mechta-Grigoriou F, Peyron JF, Trichet V, Herault O. Deciphering Tumor Niches: Lessons From Solid and Hematological Malignancies. Front Immunol 2021; 12:766275. [PMID: 34858421 PMCID: PMC8631445 DOI: 10.3389/fimmu.2021.766275] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Knowledge about the hematopoietic niche has evolved considerably in recent years, in particular through in vitro analyzes, mouse models and the use of xenografts. Its complexity in the human bone marrow, in particular in a context of hematological malignancy, is more difficult to decipher by these strategies and could benefit from the knowledge acquired on the niches of solid tumors. Indeed, some common features can be suspected, since the bone marrow is a frequent site of solid tumor metastases. Recent research on solid tumors has provided very interesting information on the interactions between tumoral cells and their microenvironment, composed notably of mesenchymal, endothelial and immune cells. This review thus focuses on recent discoveries on tumor niches that could help in understanding hematopoietic niches, with special attention to 4 particular points: i) the heterogeneity of carcinoma/cancer-associated fibroblasts (CAFs) and mesenchymal stem/stromal cells (MSCs), ii) niche cytokines and chemokines, iii) the energy/oxidative metabolism and communication, especially mitochondrial transfer, and iv) the vascular niche through angiogenesis and endothelial plasticity. This review highlights actors and/or pathways of the microenvironment broadly involved in cancer processes. This opens avenues for innovative therapeutic opportunities targeting not only cancer stem cells but also their regulatory tumor niche(s), in order to improve current antitumor therapies.
Collapse
Affiliation(s)
- Stéphane J C Mancini
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM UMR1236, Rennes 1 University, Etablissement Français du Sang Bretagne, Rennes, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France
| | - Karl Balabanian
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Saint-Louis Research Institute, University of Paris, EMiLy, INSERM U1160, Paris, France.,The Organization for Partnerships in Leukemia (OPALE) Carnot Institute, The Organization for Partnerships in Leukemia, Paris, France
| | - Isabelle Corre
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France.,Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), Signaling in Oncogenesis Angiogenesis and Permeability (SOAP), INSERM UMR1232, Centre National de la Recherche scientifique (CNRS) ERL600, Université de Nantes, Nantes, France
| | - Julie Gavard
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France.,Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), Signaling in Oncogenesis Angiogenesis and Permeability (SOAP), INSERM UMR1232, Centre National de la Recherche scientifique (CNRS) ERL600, Université de Nantes, Nantes, France.,Integrated Center for Oncology, St. Herblain, France
| | - Gwendal Lazennec
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Centre National de la Recherche scientifique (CNRS) UMR9005, SYS2DIAG-ALCEDIAG, Montpellier, France
| | - Marie-Caroline Le Bousse-Kerdilès
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM UMRS-MD1197, Paris-Saclay University, Paul-Brousse Hospital, Villejuif, France
| | - Fawzia Louache
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM UMRS-MD1197, Paris-Saclay University, Paul-Brousse Hospital, Villejuif, France
| | - Véronique Maguer-Satta
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancer Research Center of Lyon (CRCL), CNRS UMR5286, INSERM U1052, Lyon 1 university, Lean Bérard Center, Lyon, France
| | - Nathalie M Mazure
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM U1065, C3M, University of Côte d'Azur (UCA), Nice, France
| | - Fatima Mechta-Grigoriou
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Stress and Cancer Laboratory, Institut Curie, INSERM U830, Paris Sciences et Lettres (PSL) Research University, Team Babelized Ligue Nationale Contre le Cancer (LNCC), Paris, France
| | - Jean-François Peyron
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM U1065, C3M, University of Côte d'Azur (UCA), Nice, France
| | - Valérie Trichet
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France.,INSERM UMR1238 Phy-Os, Université de Nantes, Nantes, France
| | - Olivier Herault
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France.,The Organization for Partnerships in Leukemia (OPALE) Carnot Institute, The Organization for Partnerships in Leukemia, Paris, France.,Centre National de la Recherche scientifique (CNRS) ERL7001 LNOx, EA7501, Tours University, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| |
Collapse
|
29
|
Ahmed HMM, Nimmagadda SC, Al-Matary YS, Fiori M, May T, Frank D, Patnana PK, Récher C, Schliemann C, Mikesch JH, Koenig T, Rosenbauer F, Hartmann W, Tuckermann J, Dührsen U, Lanying W, Dugas M, Opalka B, Lenz G, Khandanpour C. Dexamethasone-mediated inhibition of Notch signalling blocks the interaction of leukaemia and mesenchymal stromal cells. Br J Haematol 2021; 196:995-1006. [PMID: 34792186 DOI: 10.1111/bjh.17940] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/06/2021] [Accepted: 10/21/2021] [Indexed: 11/30/2022]
Abstract
Acute myeloid leukaemia (AML) is a haematological malignancy characterized by a poor prognosis. Bone marrow mesenchymal stromal cells (BM MSCs) support leukaemic cells in preventing chemotherapy-induced apoptosis. This encouraged us to investigate leukaemia-BM niche-associated signalling and to identify signalling cascades supporting the interaction of leukaemic cells and BM MSC. Our study demonstrated functional differences between MSCs originating from leukaemic (AML MSCs) and healthy donors (HD MSCs). The direct interaction of leukaemic and AML MSCs was indispensable in influencing AML cell proliferation. We further identified an important role for Notch expression and its activation in AML MSCs contributing to the enhanced proliferation of AML cells. Supporting this observation, overexpression of the intracellular Notch domain (Notch ICN) in AML MSCs enhanced AML cells' proliferation. From a therapeutic point of view, dexamethasone treatment impeded Notch signalling in AML MSCs resulting in reduced AML cell proliferation. Concurrent with our data, Notch inhibitors had only a marginal effect on leukaemic cells alone but strongly influenced Notch signalling in AML MSCs and abrogated their cytoprotective function on AML cells. In vivo, dexamethasone treatment impeded Notch signalling in AML MSCs leading to a reduced number of AML MSCs and improved survival of leukaemic mice. In summary, targeting the interaction of leukaemic cells and AML MSCs using dexamethasone or Notch inhibitors might further improve treatment outcomes in AML patients.
Collapse
Affiliation(s)
| | - Subbaiah Chary Nimmagadda
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Yahya S Al-Matary
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany.,Department of Hematology and Stem Cell Transplantation, West German Cancer Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Maren Fiori
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany.,Department of Hematology and Stem Cell Transplantation, West German Cancer Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Daria Frank
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany.,Department of Hematology and Stem Cell Transplantation, West German Cancer Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Pradeep Kumar Patnana
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany.,Department of Hematology and Stem Cell Transplantation, West German Cancer Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christian Récher
- CHU de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Christoph Schliemann
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Jan-Henrik Mikesch
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Thorsten Koenig
- Institute of Molecular Tumor Biology, Faculty of Medicine, University of Muenster, Muenster, Germany
| | - Frank Rosenbauer
- Institute of Molecular Tumor Biology, Faculty of Medicine, University of Muenster, Muenster, Germany
| | - Wolfgang Hartmann
- Institute of Pathology, University Hospital Muenster, Muenster, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Ulrich Dührsen
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Wei Lanying
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany.,Institute of Medical Informatics, University Hospital Muenster, Muenster, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University Hospital Muenster, Muenster, Germany.,Institute of Medical Informatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Bertram Opalka
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center Essen, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Cyrus Khandanpour
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
30
|
Giri AK, Ianevski A. High-throughput screening for drug discovery targeting the cancer cell-microenvironment interactions in hematological cancers. Expert Opin Drug Discov 2021; 17:181-190. [PMID: 34743621 DOI: 10.1080/17460441.2022.1991306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION The interactions between leukemic blasts and cells within the bone marrow environment affect oncogenesis, cancer stem cell survival, as well as drug resistance in hematological cancers. The importance of this interaction is increasingly being recognized as a potentially important target for future drug discoveries and developments. Recent innovations in the high throughput drug screening-related technologies, novel ex-vivo disease-models, and freely available machine-learning algorithms are advancing the drug discovery process by targeting earlier undruggable proteins, complex pathways, as well as physical interactions (e.g. leukemic cell-bone microenvironment interaction). AREA COVERED In this review, the authors discuss the recent methodological advancements and existing challenges to target specialized hematopoietic niches within the bone marrow during leukemia and suggest how such methods can be used to identify drugs targeting leukemic cell-bone microenvironment interactions. EXPERT OPINION The recent development in cell-cell communication scoring technology and culture conditions can speed up the drug discovery by targeting the cell-microenvironment interaction. However, to accelerate this process, collecting clinical-relevant patient tissues, developing culture model systems, and implementing computational algorithms, especially trained to predict drugs and their combination targeting the cancer cell-bone microenvironment interaction are needed.
Collapse
Affiliation(s)
- Anil K Giri
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Aleksander Ianevski
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
31
|
Chen Y, Zou J, Cheng F, Li W. Treatment-Free Remission in Chronic Myeloid Leukemia and New Approaches by Targeting Leukemia Stem Cells. Front Oncol 2021; 11:769730. [PMID: 34778088 PMCID: PMC8581243 DOI: 10.3389/fonc.2021.769730] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/11/2021] [Indexed: 12/11/2022] Open
Abstract
The therapeutic landscape for chronic myeloid leukemia (CML) has improved significantly with the approval of tyrosine kinase inhibitors (TKIs) for therapeutic use. Most patients with optimal responses to TKIs can have a normal life expectancy. Treatment-free remission (TFR) after discontinuing TKI has increasingly become a new goal for CML treatment. However, TKI only "control" CML, and relapse after discontinuation has become a key factor hindering patient access to attempt TFR. In this study, we reviewed studies on TKI discontinuation, including both first and second-generation TKI. We also reviewed predictors of relapse, new monitoring methods, and strategies targeting leukemic stem cells.
Collapse
Affiliation(s)
| | | | | | - Weiming Li
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
32
|
Bolandi SM, Pakjoo M, Beigi P, Kiani M, Allahgholipour A, Goudarzi N, Khorashad JS, Eiring AM. A Role for the Bone Marrow Microenvironment in Drug Resistance of Acute Myeloid Leukemia. Cells 2021; 10:2833. [PMID: 34831055 PMCID: PMC8616250 DOI: 10.3390/cells10112833] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with a poor prognosis and remarkable resistance to chemotherapeutic agents. Understanding resistance mechanisms against currently available drugs helps to recognize the therapeutic obstacles. Various mechanisms of resistance to chemotherapy or targeted inhibitors have been described for AML cells, including a role for the bone marrow niche in both the initiation and persistence of the disease, and in drug resistance of the leukemic stem cell (LSC) population. The BM niche supports LSC survival through direct and indirect interactions among the stromal cells, hematopoietic stem/progenitor cells, and leukemic cells. Additionally, the BM niche mediates changes in metabolic and signal pathway activation due to the acquisition of new mutations or selection and expansion of a minor clone. This review briefly discusses the role of the BM microenvironment and metabolic pathways in resistance to therapy, as discovered through AML clinical studies or cell line and animal models.
Collapse
Affiliation(s)
- Seyed Mohammadreza Bolandi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Mahdi Pakjoo
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Peyman Beigi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; (M.P.); (P.B.)
| | - Mohammad Kiani
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Ali Allahgholipour
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran; (M.K.); (A.A.)
| | - Negar Goudarzi
- Department of Immunology, Razi Vaccine and Sera Research Institute, Karaj, Iran; (S.M.B.); (N.G.)
| | - Jamshid S. Khorashad
- Centre for Haematology, Hammersmith Hospital, Imperial College London, London W12 0HS, UK;
| | - Anna M. Eiring
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center at El Paso, El Paso, TX 79905, USA
| |
Collapse
|
33
|
Kerr MWA, Magalhães-Gama F, Ibiapina HNS, Hanna FSA, Xabregas LA, Alves EB, Pimentel JPD, Carvalho MPSS, Tarragô AM, Teixeira-Carvalho A, Martins-Filho OA, da Costa AG, Malheiro A. Bone Marrow Soluble Immunological Mediators as Clinical Prognosis Biomarkers in B-Cell Acute Lymphoblastic Leukemia Patients Undergoing Induction Therapy. Front Oncol 2021; 11:696032. [PMID: 34646761 PMCID: PMC8503185 DOI: 10.3389/fonc.2021.696032] [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: 04/16/2021] [Accepted: 08/25/2021] [Indexed: 11/29/2022] Open
Abstract
Different factors are used as predictors of unfavorable clinical outcomes in B-Cell Acute Lymphoblastic Leukemia (B-ALL) patients. However, new prognostic markers are needed in order to allow treatment to be more accurate, providing better results and an improved quality of life. In the present study, we have characterized the profile of bone marrow soluble mediators as possible biomarkers for risk group stratification and minimal residual disease (MRD) detection during induction therapy. The study featured 47 newly-diagnosed B-cell acute lymphoblastic leukemia (B-ALL) patients that were categorized into subgroups during induction therapy according to risk stratification at day 15 [Low Risk (LR), Low Risk increasing to High Risk (LR→HR) and High Risk (HR)] and the MRD detection on day 35 (MRD(-) and MRD(+)). Soluble immunological mediators (CXCL8, CCL2, CXCL9, CCL5, CXCL10, IL-1β, IL-6, TNF, IFN-γ, IL-17A, IL-4, IL-5, IL-10 and IL-2) were quantified by cytometric bead array and ELISA. Our findings demonstrated that increased levels of CCL5, IFN-γ and IL-2 at baseline appeared as putative candidates of good prognosis in LR and MRD(-) subgroups, while CCL2 was identified as a consistent late biomarker associated with poor prognosis, which was observed on D35 in HR and MRD(+) subgroups. Furthermore, apparently controversial data regarding IL-17A and TNF did not allow the definition of these molecules as either positive or negative biomarkers. These results contribute to the search for novel prognostic indicators, and indicate the potential of bone marrow soluble mediators in prognosis and follow-up of B-ALL patients during induction therapy.
Collapse
Affiliation(s)
- Marlon Wendell Athaydes Kerr
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Fábio Magalhães-Gama
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil.,Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, Brazil.,Programa de Pós-Graduação em Ciências da Saúde, Instituto René Rachou - Fundação Oswaldo Cruz (FIOCRUZ) Minas, Belo Horizonte, Brazil.,Grupo Integrado de Pesquisas em Biomarcadores de Diagnóstico e Monitoração, Instituto René Rachou - FIOCRUZ Minas, Belo Horizonte, Brazil
| | - Hiochelson Najibe Santos Ibiapina
- Programa de Pós-Graduação em Medicina Tropical, UEA, Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil
| | - Fabíola Silva Alves Hanna
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil.,Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Lilyane Amorim Xabregas
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Eliana Brasil Alves
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - João Paulo Diniz Pimentel
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Maria Perpétuo Socorro Sampaio Carvalho
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Andréa Monteiro Tarragô
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil.,Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Andréa Teixeira-Carvalho
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Programa de Pós-Graduação em Ciências da Saúde, Instituto René Rachou - Fundação Oswaldo Cruz (FIOCRUZ) Minas, Belo Horizonte, Brazil.,Grupo Integrado de Pesquisas em Biomarcadores de Diagnóstico e Monitoração, Instituto René Rachou - FIOCRUZ Minas, Belo Horizonte, Brazil
| | - Olindo Assis Martins-Filho
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Programa de Pós-Graduação em Ciências da Saúde, Instituto René Rachou - Fundação Oswaldo Cruz (FIOCRUZ) Minas, Belo Horizonte, Brazil.,Grupo Integrado de Pesquisas em Biomarcadores de Diagnóstico e Monitoração, Instituto René Rachou - FIOCRUZ Minas, Belo Horizonte, Brazil
| | - Allyson Guimarães da Costa
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil.,Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, Brazil.,Programa de Pós-Graduação em Medicina Tropical, UEA, Manaus, Brazil.,Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, Brazil.,Escola de Enfermagem de Manaus, UFAM, Manaus, Brazil
| | - Adriana Malheiro
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil.,Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil.,Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| |
Collapse
|
34
|
Gynn LE, Anderson E, Robinson G, Wexler SA, Upstill-Goddard G, Cox C, May JE. Primary mesenchymal stromal cells in co-culture with leukaemic HL-60 cells are sensitised to cytarabine-induced genotoxicity, whilst leukaemic cells are protected. Mutagenesis 2021; 36:419-428. [PMID: 34505878 PMCID: PMC8633936 DOI: 10.1093/mutage/geab033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 09/09/2021] [Indexed: 12/27/2022] Open
Abstract
Tumour microenvironments are hallmarked in many cancer types. In haematological malignancies, bone marrow (BM) mesenchymal stromal cells (MSC) protect malignant cells from drug-induced cytotoxicity. However, less is known about malignant impact on supportive stroma. Notably, it is unknown whether these interactions alter long-term genotoxic damage in either direction. The nucleoside analogue cytarabine (ara-C), common in haematological therapies, remains the most effective agent for acute myeloid leukaemia, yet one third of patients develop resistance. This study aimed to evaluate the bidirectional effect of MSC and malignant cell co-culture on ara-C genotoxicity modulation. Primary MSC, isolated from patient BM aspirates for haematological investigations, and malignant haematopoietic cells (leukaemic HL-60) were co-cultured using trans-well inserts, prior to treatment with physiological dose ara-C. Co-culture genotoxic effects were assessed by micronucleus and alkaline comet assays. Patient BM cells from chemotherapy-treated patients had reduced ex vivo survival (P = 0.0049) and increased genotoxicity (P = 0.3172) than untreated patients. It was shown for the first time that HL-60 were protected by MSC from ara-C-induced genotoxicity, with reduced MN incidence in co-culture as compared to mono-culture (P = 0.0068). Comet tail intensity also significantly increased in ara-C-treated MSC with HL-60 influence (P = 0.0308). MSC sensitisation to ara-C genotoxicity was also demonstrated following co-culture with HL60 (P = 0.0116), which showed significantly greater sensitisation when MSC-HL-60 co-cultures were exposed to ara-C (P = 0.0409). This study shows for the first time that malignant HSC and MSC bidirectionally modulate genotoxicity, providing grounding for future research identifying mechanisms of altered genotoxicity in leukaemic microenvironments. MSC retain long-term genotoxic and functional damage following chemotherapy exposure. Understanding the interactions perpetuating such damage may inform modifications to reduce therapy-related complications, such as secondary malignancies and BM failure.
Collapse
Affiliation(s)
- Liana E Gynn
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Elizabeth Anderson
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Gareth Robinson
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Sarah A Wexler
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.,Royal United Hospitals Bath NHS Foundation Trust, Bath, BA1 3NG, UK
| | - Gillian Upstill-Goddard
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.,Royal United Hospitals Bath NHS Foundation Trust, Bath, BA1 3NG, UK
| | - Christine Cox
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.,Royal United Hospitals Bath NHS Foundation Trust, Bath, BA1 3NG, UK
| | - Jennifer E May
- Centre for Research in Biosciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| |
Collapse
|
35
|
Woods K, Guezguez B. Dynamic Changes of the Bone Marrow Niche: Mesenchymal Stromal Cells and Their Progeny During Aging and Leukemia. Front Cell Dev Biol 2021; 9:714716. [PMID: 34447754 PMCID: PMC8383146 DOI: 10.3389/fcell.2021.714716] [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: 05/25/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are a heterogenous cell population found in a wide range of tissues in the body, known for their nutrient-producing and immunomodulatory functions. In the bone marrow (BM), these MSCs are critical for the regulation of hematopoietic stem cells (HSC) that are responsible for daily blood production and functional immunity throughout an entire organism's lifespan. Alongside other stromal cells, MSCs form a specialized microenvironment BM tissue called "niche" that tightly controls HSC self-renewal and differentiation. In addition, MSCs are crucial players in maintaining bone integrity and supply of hormonal nutrients due to their capacity to differentiate into osteoblasts and adipocytes which also contribute to cellular composition of the BM niche. However, MSCs are known to encompass a large heterogenous cell population that remains elusive and poorly defined. In this review, we focus on deciphering the BM-MSC biology through recent advances in single-cell identification of hierarchical subsets with distinct functionalities and transcriptional profiles. We also discuss the contribution of MSCs and their osteo-adipo progeny in modulating the complex direct cell-to-cell or indirect soluble factors-mediated interactions of the BM HSC niche during homeostasis, aging and myeloid malignancies. Lastly, we examine the therapeutic potential of MSCs for rejuvenation and anti-tumor remedy in clinical settings.
Collapse
Affiliation(s)
- Kevin Woods
- German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- Department of Hematology and Oncology, University Medical Center Mainz, Mainz, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Borhane Guezguez
- German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- Department of Hematology and Oncology, University Medical Center Mainz, Mainz, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
36
|
Fard MB, Atashi A, Amiri S, Kaviani S, Gholampour MA, Ajami M. Parvovirus B19 affects thrombopoietin and IL-11 gene expression in human bone marrow mesenchymal stem cells. Future Virol 2021. [DOI: 10.2217/fvl-2021-0020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: Human bone marrow mesenchymal stem cells (hBMSCs) may be infected by parvovirus B19 (B19V). hBMSCs support bone marrow hematopoiesis by producing stromal cells, secretion of cytokines and growth factors, etc. Because of the lifetime persistent infection of the virus in healthy individual’s bone marrow, this study aims to evaluate B19V effects on hBMSCs gene expression of some crucial hematopoietic cytokines. Materials & methods: hBMSCs were transfected with pHI0 plasmid containing the B19V genome. The quantitative mRNA expression of target genes was evaluated 24 h after transfection. Results: Our findings demonstrated a significant increase in expression levels of IL-11 and TPO (p < 0.05). Conclusion: We concluded that alteration in the gene expressions in B19V-infected hBMSCs might have significant effects on the bone marrow microenvironment as well as hematopoiesis.
Collapse
Affiliation(s)
- Mahin Behzadi Fard
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Amir Atashi
- Stem Cell & Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Shahin Amiri
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Saeid Kaviani
- Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Mansoureh Ajami
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
37
|
Leukemia-Induced Cellular Senescence and Stemness Alterations in Mesenchymal Stem Cells Are Reversible upon Withdrawal of B-Cell Acute Lymphoblastic Leukemia Cells. Int J Mol Sci 2021; 22:ijms22158166. [PMID: 34360930 PMCID: PMC8348535 DOI: 10.3390/ijms22158166] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/12/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022] Open
Abstract
Leukemic cell growth in the bone marrow (BM) induces a very stressful condition. Mesenchymal stem cells (MSC), a key component of this BM niche, are affected in several ways with unfavorable consequences on hematopoietic stem cells favoring leukemic cells. These alterations in MSC during B-cell acute lymphoblastic leukemia (B-ALL) have not been fully studied. In this work, we have compared the modifications that occur in an in vitro leukemic niche (LN) with those observed in MSC isolated from B-ALL patients. MSC in this LN niche showed features of a senescence process, i.e., altered morphology, increased senescence-associated β-Galactosidase (SA-βGAL) activity, and upregulation of p53 and p21 (without p16 expression), cell-cycle arrest, reduced clonogenicity, and some moderated changes in stemness properties. Importantly, almost all of these features were found in MSC isolated from B-ALL patients. These alterations rendered B-ALL cells susceptible to the chemotherapeutic agent dexamethasone. The senescent process seems to be transient since when leukemic cells are removed, normal MSC morphology is re-established, SA-βGAL expression is diminished, and MSC are capable of re-entering cell cycle. In addition, few cells showed low γH2AX phosphorylation that was reduced to basal levels upon cultivation. The reversibility of the senescent process in MSC must impinge important biological and clinical significance depending on cell interactions in the bone marrow at different stages of disease progression in B-ALL.
Collapse
|
38
|
Kapor S, Santibanez JF. Myeloid-Derived Suppressor Cells and Mesenchymal Stem/Stromal Cells in Myeloid Malignancies. J Clin Med 2021; 10:2788. [PMID: 34202907 PMCID: PMC8268878 DOI: 10.3390/jcm10132788] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/14/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022] Open
Abstract
Myeloid malignancies arise from an altered hematopoietic stem cell and mainly comprise acute myeloid leukemia, myelodysplastic syndromes, myeloproliferative malignancies, and chronic myelomonocytic leukemia. Myeloid neoplastic leukemic cells may influence the growth and differentiation of other hematopoietic cell lineages in peripheral blood and bone marrow. Myeloid-derived suppressor cells (MDSCs) and mesenchymal stromal cells (MSCs) display immunoregulatory properties by controlling the innate and adaptive immune systems that may induce a tolerant and supportive microenvironment for neoplasm development. This review analyzes the main features of MDSCs and MSCs in myeloid malignancies. The number of MDSCs is elevated in myeloid malignancies exhibiting high immunosuppressive capacities, whereas MSCs, in addition to their immunosuppression contribution, regulate myeloid leukemia cell proliferation, apoptosis, and chemotherapy resistance. Moreover, MSCs may promote MDSC expansion, which may mutually contribute to the creation of an immuno-tolerant neoplasm microenvironment. Understanding the implication of MDSCs and MSCs in myeloid malignancies may favor their potential use in immunotherapeutic strategies.
Collapse
Affiliation(s)
- Suncica Kapor
- Clinical Hospital Center “Dr Dragisa Misovic-Dedinje”, Department of Hematology, University of Belgrade, 11000 Belgrade, Serbia
| | - Juan F. Santibanez
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia;
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, 8370993 Santiago, Chile
| |
Collapse
|
39
|
Kapor S, Santibanez JF. Myeloid-Derived Suppressor Cells and Mesenchymal Stem/Stromal Cells in Myeloid Malignancies. J Clin Med 2021. [PMID: 34202907 DOI: 10.3390/jcm10132788.pmid:34202907;pmcid:pmc8268878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Myeloid malignancies arise from an altered hematopoietic stem cell and mainly comprise acute myeloid leukemia, myelodysplastic syndromes, myeloproliferative malignancies, and chronic myelomonocytic leukemia. Myeloid neoplastic leukemic cells may influence the growth and differentiation of other hematopoietic cell lineages in peripheral blood and bone marrow. Myeloid-derived suppressor cells (MDSCs) and mesenchymal stromal cells (MSCs) display immunoregulatory properties by controlling the innate and adaptive immune systems that may induce a tolerant and supportive microenvironment for neoplasm development. This review analyzes the main features of MDSCs and MSCs in myeloid malignancies. The number of MDSCs is elevated in myeloid malignancies exhibiting high immunosuppressive capacities, whereas MSCs, in addition to their immunosuppression contribution, regulate myeloid leukemia cell proliferation, apoptosis, and chemotherapy resistance. Moreover, MSCs may promote MDSC expansion, which may mutually contribute to the creation of an immuno-tolerant neoplasm microenvironment. Understanding the implication of MDSCs and MSCs in myeloid malignancies may favor their potential use in immunotherapeutic strategies.
Collapse
Affiliation(s)
- Suncica Kapor
- Clinical Hospital Center "Dr Dragisa Misovic-Dedinje", Department of Hematology, University of Belgrade, 11000 Belgrade, Serbia
| | - Juan F Santibanez
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, 11000 Belgrade, Serbia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, 8370993 Santiago, Chile
| |
Collapse
|
40
|
Michelozzi IM, Kirtsios E, Giustacchini A. Driving CAR T Stem Cell Targeting in Acute Myeloid Leukemia: The Roads to Success. Cancers (Basel) 2021; 13:2816. [PMID: 34198742 PMCID: PMC8201025 DOI: 10.3390/cancers13112816] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 12/14/2022] Open
Abstract
Current treatment outcome for acute myeloid leukemia (AML) patients is unsatisfactory and characterized by high rates of relapse and poor overall survival. Increasing evidence points to a crucial role of leukemic stem cells (LSC) and the bone marrow (BM) leukemic niche, in which they reside, in AML evolution and chemoresistance. Thus, future strategies aiming at improving AML therapeutic protocols are likely to be directed against LSC and their niche. Chimeric antigen receptor (CAR) T-cells have been extremely successful in the treatment of relapsed/refractory acute lymphoblastic leukemia and B-cell non-Hodgkin lymphoma and comparable results in AML are highly desirable. At present, we are at the dawn of CAR T-cell application in AML, with several preclinical studies and few early phase clinical trials. However, the lack of leukemia-specific targets and the genetic and phenotypic heterogeneity of the disease combined with the leukemia-induced remodeling of the BM microenvironment are limiting CAR T-cell exploitation in AML. Here, we reviewed AML-LSC and AML-BM niche features in the context of their therapeutic targeting using CAR T-cells. We summarized recent progress in CAR T-cell application to the treatment of AML, and we discussed the remaining therapeutic challenges and promising novel strategies to overcome them.
Collapse
Affiliation(s)
- Ilaria M. Michelozzi
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, London WC1N 1DZ, UK;
| | | | - Alice Giustacchini
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, London WC1N 1DZ, UK;
| |
Collapse
|
41
|
Acute myeloid leukemia-induced remodeling of the human bone marrow niche predicts clinical outcome. Blood Adv 2021; 4:5257-5268. [PMID: 33108453 DOI: 10.1182/bloodadvances.2020001808] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022] Open
Abstract
Murine models of myeloid neoplasia show how leukemia infiltration alters the hematopoietic stem cell (HSC) niche to reinforce malignancy at the expense of healthy hematopoiesis. However, little is known about the bone marrow architecture in humans and its impact on clinical outcome. Here, we dissect the bone marrow niche in patients with acute myeloid leukemia (AML) at first diagnosis. We combined immunohistochemical stainings with global gene expression analyses from these AML patients and correlated them with clinical features. Mesenchymal stem and progenitor cells (MSPCs) lost quiescence and significantly expanded in the bone marrow of AML patients. Strikingly, their HSC- and niche-regulating capacities were impaired with significant inhibition of osteogenesis and bone formation in a cell contact-dependent manner through inhibition of cytoplasmic β-catenin. Assessment of bone metabolism by quantifying peripheral blood osteocalcin levels revealed 30% lower expression in AML patients at first diagnosis than in non-leukemic donors. Furthermore, patients with osteocalcin levels ≤11 ng/mL showed inferior overall survival with a 1-year survival rate of 38.7% whereas patients with higher osteocalcin levels reached a survival rate of 66.8%. These novel insights into the human AML bone marrow microenvironment help translate findings from preclinical models and detect new targets which might pave the way for niche-targeted therapies in AML patients.
Collapse
|
42
|
Sendker S, Waack K, Reinhardt D. Far from Health: The Bone Marrow Microenvironment in AML, A Leukemia Supportive Shelter. CHILDREN (BASEL, SWITZERLAND) 2021; 8:371. [PMID: 34066861 PMCID: PMC8150304 DOI: 10.3390/children8050371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/28/2022]
Abstract
Acute myeloid leukemia (AML) is the second most common leukemia among children. Although significant progress in AML therapy has been achieved, treatment failure is still associated with poor prognosis, emphasizing the need for novel, innovative therapeutic approaches. To address this major obstacle, extensive knowledge about leukemogenesis and the complex interplay between leukemic cells and their microenvironment is required. The tremendous role of this bone marrow microenvironment in providing a supportive and protective shelter for leukemic cells, leading to disease development, progression, and relapse, has been emphasized by recent research. It has been revealed that the interplay between leukemic cells and surrounding cellular as well as non-cellular components is critical in the process of leukemogenesis. In this review, we provide a comprehensive overview of recently gained knowledge about the importance of the microenvironment in AML whilst focusing on promising future therapeutic targets. In this context, we describe ongoing clinical trials and future challenges for the development of targeted therapies for AML.
Collapse
Affiliation(s)
| | | | - Dirk Reinhardt
- Department of Pediatric Hematology and Oncology, Clinic of Pediatrics III, Essen University Hospital, 45147 Essen, Germany; (S.S.); (K.W.)
| |
Collapse
|
43
|
Nasal Turbinate Mesenchymal Stromal Cells Preserve Characteristics of Their Neural Crest Origin and Exert Distinct Paracrine Activity. J Clin Med 2021; 10:jcm10081792. [PMID: 33924095 PMCID: PMC8074274 DOI: 10.3390/jcm10081792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/06/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023] Open
Abstract
The sources of mesenchymal stromal cells (MSCs) for cell therapy trials are expanding, increasing the need for their characterization. Here, we characterized multi-donor, turbinate-derived MSCs (TB-MSCs) that develop from the neural crest, and compared them to bone marrow-derived MSCs (BM-MSCs). TB-MSCs had higher proliferation potential and higher self-renewal of colony forming cells, but lower potential for multi-lineage differentiation than BM-MSCs. TB-MSCs expressed higher levels of neural crest markers and lower levels of pericyte-specific markers. These neural crest-like properties of TB-MSCs were reflected by their propensity to differentiate into neuronal cells and proliferative response to nerve growth factors. Proteomics (LC-MS/MS) analysis revealed a distinct secretome profile of TB-MSCs compared to BM and adipose tissue-derived MSCs, exhibiting enrichments of factors for cell-extracellular matrix interaction and neurogenic signaling. However, TB-MSCs and BM-MSCs exhibited comparable suppressive effects on the allo-immune response and comparable stimulatory effects on hematopoietic stem cell self-renewal. In contrast, TB-MSCs stimulated growth and metastasis of breast cancer cells more than BM-MSCs. Altogether, our multi-donor characterization of TB-MSCs reveals distinct cell autonomous and paracrine properties, reflecting their unique developmental origin. These findings support using TB-MSCs as an alternative source of MSCs with distinct biological characteristics for optimal applications in cell therapy.
Collapse
|
44
|
Lee HR, Yang SJ, Choi HK, Kim JA, Oh IH. The Chromatin Remodeling Complex CHD1 Regulates the Primitive State of Mesenchymal Stromal Cells to Control Their Stem Cell Supporting Activity. Stem Cells Dev 2021; 30:363-373. [PMID: 33593142 DOI: 10.1089/scd.2020.0166] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The primitive state (stemness) of mesenchymal stromal cells (MSCs) is responsible for supporting the function of tissue-specific stem cells to regenerate damaged tissues. However, molecular mechanisms regulating the stemness of MSCs remain unknown. In this study, we found that the primitive state of MSCs is hierarchically regulated by the expression levels of the chromatin remodeling complex, CHD1, with CHD1 expression levels higher in the undifferentiated state, and decreasing upon MSC differentiation. Consistently, CHD1 expression levels decrease during progressive loss of clonogenic progenitors (CFU-F) induced by passage cultures. Moreover, knockdown (KD) of CHD1 decreased CFU-F frequency, whereas CHD1 overexpression increased it. In addition, the expression of stem cell-specific genes was down- or upregulated upon KD or overexpression of CHD1, respectively, accompanied by associated changes in chromatin condensation. Importantly, altering CHD1 expression levels affected the ability of MSCs to support the self-renewing expansion of hematopoietic stem cells (HSCs). Furthermore, CHD1 levels were significantly decreased in MSCs from acute myeloid leukemia or aplastic anemia patients, where CFU-F and HSC-supporting activities are lost. Altogether, these findings show that chromatin remodeling by CHD1 is a molecular parameter that influences the primitive state of MSCs and their stem cell-supporting activity, which controls tissue regeneration.
Collapse
Affiliation(s)
- Hae-Ri Lee
- Catholic High-Performance Cell Therapy Center, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Jip Yang
- Catholic High-Performance Cell Therapy Center, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun-Kyung Choi
- Catholic High-Performance Cell Therapy Center, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin-A Kim
- Catholic High-Performance Cell Therapy Center, The Catholic University of Korea, Seoul, Republic of Korea
| | - Il-Hoan Oh
- Catholic High-Performance Cell Therapy Center, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
45
|
Pievani A, Donsante S, Tomasoni C, Corsi A, Dazzi F, Biondi A, Riminucci M, Serafini M. Acute myeloid leukemia shapes the bone marrow stromal niche in vivo. Haematologica 2021; 106:865-870. [PMID: 32381570 PMCID: PMC7928008 DOI: 10.3324/haematol.2020.247205] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Indexed: 01/04/2023] Open
Affiliation(s)
- Alice Pievani
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | | | - Chiara Tomasoni
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Francesco Dazzi
- Department of Hemato-Oncology, Rayne Institute, King’s College London, London, UK
| | - Andrea Biondi
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
- Department of Pediatrics, Fondazione MBBM/San Gerardo Hospital, Monza, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Marta Serafini
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
46
|
Zhang H, Zhang R, Zheng X, Sun M, Fan J, Fang C, Tian X, Zheng H. BACH2-mediated FOS confers cytarabine resistance via stromal microenvironment alterations in pediatric ALL. Cancer Sci 2021; 112:1235-1250. [PMID: 33393145 PMCID: PMC7935781 DOI: 10.1111/cas.14792] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/30/2020] [Accepted: 12/30/2020] [Indexed: 12/24/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is an aggressive hematological cancer that mainly affects children. Relapse and chemoresistance result in treatment failure, underlining the need for improved therapies. BTB and CNC homology 2 (BACH2) is a lymphoid-specific transcription repressor recognized as a tumor suppressor in lymphomas, but little is known about its function and regulatory network in pediatric ALL (p-ALL). Herein, we found aberrant BACH2 expression at new diagnosis not only facilitated risk stratification of p-ALL but also served as a sensitive predictor of early treatment response and clinical outcome. Silencing BACH2 in ALL cells increased cell proliferation and accelerated cell cycle progression. BACH2 blockade also promoted cell adhesion to bone marrow stromal cells and conferred cytarabine (Ara-C)-resistant properties to leukemia cells by altering stromal microenvironment. Strikingly, we identified FOS, a transcriptional activator competing with BACH2, as a novel downstream target repressed by BACH2. Blocking FOS by chemical compounds enhanced the effect of Ara-C treatment in both primary p-ALL cells and pre-B-ALL-driven leukemia xenografts and prolonged the survival of tumor-bearing mice. These data highlight an interconnected network of BACH2-FOS, disruption of which could render current chemotherapies more effective and offer a promising therapeutic strategy to overcome Ara-C resistance in p-ALL.
Collapse
Affiliation(s)
- Han Zhang
- Institute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
| | - Ruidong Zhang
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics (Capital Medical University)Key Laboratory of Major Diseases in ChildrenMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalNational Center for Children’s HealthCapital Medical UniversityBeijingChina
| | - Xueling Zheng
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics (Capital Medical University)Key Laboratory of Major Diseases in ChildrenMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalNational Center for Children’s HealthCapital Medical UniversityBeijingChina
| | - Ming Sun
- Institute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeKunmingChina
| | - Jia Fan
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics (Capital Medical University)Key Laboratory of Major Diseases in ChildrenMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalNational Center for Children’s HealthCapital Medical UniversityBeijingChina
| | - Chunlian Fang
- Department of Hematology and OncologyKunming Children’s Hospital (Children’s Hospital of Kunming Medical University, Yunnan Children’s Medical Center)KunmingChina
| | - Xin Tian
- Department of Hematology and OncologyKunming Children’s Hospital (Children’s Hospital of Kunming Medical University, Yunnan Children’s Medical Center)KunmingChina
| | - Huyong Zheng
- Beijing Key Laboratory of Pediatric Hematology OncologyNational Key Discipline of Pediatrics (Capital Medical University)Key Laboratory of Major Diseases in ChildrenMinistry of EducationHematology Oncology CenterBeijing Children’s HospitalNational Center for Children’s HealthCapital Medical UniversityBeijingChina
| |
Collapse
|
47
|
Yamaguchi T, Kawamoto E, Gaowa A, Park EJ, Shimaoka M. Remodeling of Bone Marrow Niches and Roles of Exosomes in Leukemia. Int J Mol Sci 2021; 22:ijms22041881. [PMID: 33668652 PMCID: PMC7918833 DOI: 10.3390/ijms22041881] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/23/2021] [Accepted: 02/11/2021] [Indexed: 12/15/2022] Open
Abstract
Leukemia is a hematological malignancy that originates from hematopoietic stem cells in the bone marrow. Significant progress has made in understanding its pathogensis and in establishing chemotherapy and hematopoietic stem cell transplantation therapy (HSCT). However, while the successive development of new therapies, such as molecular-targeted therapy and immunotherapy, have resulted in remarkable advances, the fact remains that some patients still cannot be saved, and resistance to treatment and relapse are still problems that need to be solved in leukemia patients. The bone marrow (BM) niche is a microenvironment that includes hematopoietic stem cells and their supporting cells. Leukemia cells interact with bone marrow niches and modulate them, not only inducing molecular and functional changes but also switching to niches favored by leukemia cells. The latter are closely associated with leukemia progression, suppression of normal hematopoiesis, and chemotherapy resistance, which is precisely the area of ongoing study. Exosomes play an important role in cell-to-cell communication, not only with cells in close proximity but also with those more distant due to the nature of exosomal circulation via body fluids. In leukemia, exosomes play important roles in leukemogenesis, disease progression, and organ invasion, and their usefulness in the diagnosis and treatment of leukemia has recently been reported. The interaction between leukemia cell-derived exosomes and the BM microenvironment has received particular attention. Their interaction is believed to play a very important role; in addition to their diagnostic value, exosomes could serve as a marker for monitoring treatment efficacy and as an aid in overcoming drug resistance, among the many problems in leukemia patients that have yet to be overcome. In this paper, we will review bone marrow niches in leukemia, findings on leukemia-derived exosomes, and exosome-induced changes in bone marrow niches.
Collapse
Affiliation(s)
- Takanori Yamaguchi
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City, Mie 514-8507, Japan; (T.Y.); (E.K.); (A.G.); (E.J.P.)
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City, Mie 514-8507, Japan
| | - Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City, Mie 514-8507, Japan; (T.Y.); (E.K.); (A.G.); (E.J.P.)
- Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City, Mie 514-8507, Japan
| | - Arong Gaowa
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City, Mie 514-8507, Japan; (T.Y.); (E.K.); (A.G.); (E.J.P.)
| | - Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City, Mie 514-8507, Japan; (T.Y.); (E.K.); (A.G.); (E.J.P.)
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu-City, Mie 514-8507, Japan; (T.Y.); (E.K.); (A.G.); (E.J.P.)
- Correspondence: ; Tel.: +81-59-232-5036; Fax: +81-59-231-5209
| |
Collapse
|
48
|
Lee HR, Lee GY, Kim EW, Kim HJ, Lee M, Humphries RK, Oh IH. Reversible switching of leukemic cells to a drug-resistant, stem-like subset via IL-4 mediated cross-talk with mesenchymal stroma. Haematologica 2021; 107:381-392. [PMID: 33440923 PMCID: PMC8804570 DOI: 10.3324/haematol.2020.269944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Indexed: 11/13/2022] Open
Abstract
Chemoresistance of leukemic cells has largely been attributed to clonal evolution secondary to accumulating mutations. Here, we show that a subset of leukemic blasts in contact with the mesenchymal stroma undergo cellular conversion into a distinct cell type that exhibits a stem cell-like phenotype and chemoresistance. These stroma-induced changes occur in a reversible and stochastic manner driven by cross-talk, whereby stromal contact induces interleukin-4 in leukemic cells that in turn targets the mesenchymal stroma to facilitate the development of new subset. This mechanism was dependent on interleukin-4-mediated upregulation of vascular cell adhesion molecule- 1 in mesenchymal stroma, causing tight adherence of leukemic cells to mesenchymal progenitors for generation of new subsets. Together, our study reveals another class of chemoresistance in leukemic blasts via functional evolution through stromal cross-talk, and demonstrates dynamic switching of leukemic cell fates that could cause a non-homologous response to chemotherapy in concert with the patient-specific microenvironment.
Collapse
Affiliation(s)
- Hae-Ri Lee
- Catholic High-Performance Cell Therapy Center and Department of Medical Life Science, College of Medicine, The Catholic University, Seoul
| | - Ga-Young Lee
- Catholic High-Performance Cell Therapy Center and Department of Medical Life Science, College of Medicine, The Catholic University, Seoul
| | - Eung-Won Kim
- Catholic High-Performance Cell Therapy Center and Department of Medical Life Science, College of Medicine, The Catholic University, Seoul
| | - Hee-Je Kim
- Division of Hematology, Department of Internal Medicine, St Mary's Hematology Hospital, College of Medicine, The Catholic University of Korea
| | - Minho Lee
- Department of Life Science, Dongguk University-Seoul, Goyang-si, Gyeonggi-do
| | - R Keith Humphries
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada; Department of Medicine, University of British Columbia, Vancouver
| | - Il-Hoan Oh
- Catholic High-Performance Cell Therapy Center and Department of Medical Life Science, College of Medicine, The Catholic University, Seoul.
| |
Collapse
|
49
|
Takam Kamga P, Bazzoni R, Dal Collo G, Cassaro A, Tanasi I, Russignan A, Tecchio C, Krampera M. The Role of Notch and Wnt Signaling in MSC Communication in Normal and Leukemic Bone Marrow Niche. Front Cell Dev Biol 2021; 8:599276. [PMID: 33490067 PMCID: PMC7820188 DOI: 10.3389/fcell.2020.599276] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022] Open
Abstract
Notch and Wnt signaling are highly conserved intercellular communication pathways involved in developmental processes, such as hematopoiesis. Even though data from literature support a role for these two pathways in both physiological hematopoiesis and leukemia, there are still many controversies concerning the nature of their contribution. Early studies, strengthened by findings from T-cell acute lymphoblastic leukemia (T-ALL), have focused their investigation on the mutations in genes encoding for components of the pathways, with limited results except for B-cell chronic lymphocytic leukemia (CLL); in because in other leukemia the two pathways could be hyper-expressed without genetic abnormalities. As normal and malignant hematopoiesis require close and complex interactions between hematopoietic cells and specialized bone marrow (BM) niche cells, recent studies have focused on the role of Notch and Wnt signaling in the context of normal crosstalk between hematopoietic/leukemia cells and stromal components. Amongst the latter, mesenchymal stromal/stem cells (MSCs) play a pivotal role as multipotent non-hematopoietic cells capable of giving rise to most of the BM niche stromal cells, including fibroblasts, adipocytes, and osteocytes. Indeed, MSCs express and secrete a broad pattern of bioactive molecules, including Notch and Wnt molecules, that support all the phases of the hematopoiesis, including self-renewal, proliferation and differentiation. Herein, we provide an overview on recent advances on the contribution of MSC-derived Notch and Wnt signaling to hematopoiesis and leukemia development.
Collapse
Affiliation(s)
- Paul Takam Kamga
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
- EA4340-BCOH, Biomarker in Cancerology and Onco-Haematology, UVSQ, Université Paris Saclay, Boulogne-Billancourt, France
| | - Riccardo Bazzoni
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Giada Dal Collo
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Adriana Cassaro
- Hematology Unit, Department of Oncology, Niguarda Hospital, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Ilaria Tanasi
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Anna Russignan
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Cristina Tecchio
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Mauro Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
50
|
Forte D, García-Fernández M, Sánchez-Aguilera A, Stavropoulou V, Fielding C, Martín-Pérez D, López JA, Costa ASH, Tronci L, Nikitopoulou E, Barber M, Gallipoli P, Marando L, Fernández de Castillejo CL, Tzankov A, Dietmann S, Cavo M, Catani L, Curti A, Vázquez J, Frezza C, Huntly BJ, Schwaller J, Méndez-Ferrer S. Bone Marrow Mesenchymal Stem Cells Support Acute Myeloid Leukemia Bioenergetics and Enhance Antioxidant Defense and Escape from Chemotherapy. Cell Metab 2020; 32:829-843.e9. [PMID: 32966766 PMCID: PMC7658808 DOI: 10.1016/j.cmet.2020.09.001] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 05/12/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022]
Abstract
Like normal hematopoietic stem cells, leukemic stem cells depend on their bone marrow (BM) microenvironment for survival, but the underlying mechanisms remain largely unknown. We have studied the contribution of nestin+ BM mesenchymal stem cells (BMSCs) to MLL-AF9-driven acute myeloid leukemia (AML) development and chemoresistance in vivo. Unlike bulk stroma, nestin+ BMSC numbers are not reduced in AML, but their function changes to support AML cells, at the expense of non-mutated hematopoietic stem cells (HSCs). Nestin+ cell depletion delays leukemogenesis in primary AML mice and selectively decreases AML, but not normal, cells in chimeric mice. Nestin+ BMSCs support survival and chemotherapy relapse of AML through increased oxidative phosphorylation, tricarboxylic acid (TCA) cycle activity, and glutathione (GSH)-mediated antioxidant defense. Therefore, AML cells co-opt energy sources and antioxidant defense mechanisms from BMSCs to survive chemotherapy.
Collapse
Affiliation(s)
- Dorian Forte
- Wellcome-MRC Cambridge Stem Cell Institute, CB2 0AW Cambridge, UK; National Health Service Blood and Transplant, CB2 0PT Cambridge, UK; Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università degli Studi, 40138 Bologna, Italy
| | - María García-Fernández
- Wellcome-MRC Cambridge Stem Cell Institute, CB2 0AW Cambridge, UK; National Health Service Blood and Transplant, CB2 0PT Cambridge, UK
| | | | - Vaia Stavropoulou
- University Children's Hospital and Department of Biomedicine (DBM), University of Basel, 4031 Basel, Switzerland
| | - Claire Fielding
- Wellcome-MRC Cambridge Stem Cell Institute, CB2 0AW Cambridge, UK; National Health Service Blood and Transplant, CB2 0PT Cambridge, UK
| | - Daniel Martín-Pérez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Juan Antonio López
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Ana S H Costa
- MRC Cancer Unit, University of Cambridge, CB2 0XZ Cambridge, UK
| | - Laura Tronci
- MRC Cancer Unit, University of Cambridge, CB2 0XZ Cambridge, UK
| | | | - Michael Barber
- Wellcome-MRC Cambridge Stem Cell Institute, CB2 0AW Cambridge, UK
| | - Paolo Gallipoli
- Wellcome-MRC Cambridge Stem Cell Institute, CB2 0AW Cambridge, UK; Department of Haematology, University of Cambridge, CB2 0AW Cambridge, UK
| | - Ludovica Marando
- Wellcome-MRC Cambridge Stem Cell Institute, CB2 0AW Cambridge, UK; Department of Haematology, University of Cambridge, CB2 0AW Cambridge, UK
| | | | - Alexandar Tzankov
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland
| | - Sabine Dietmann
- Wellcome-MRC Cambridge Stem Cell Institute, CB2 0AW Cambridge, UK
| | - Michele Cavo
- Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università degli Studi, 40138 Bologna, Italy; Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, 40138 Bologna, Italy
| | - Lucia Catani
- Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università degli Studi, 40138 Bologna, Italy; Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, 40138 Bologna, Italy
| | - Antonio Curti
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, 40138 Bologna, Italy
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | | | - Brian J Huntly
- Wellcome-MRC Cambridge Stem Cell Institute, CB2 0AW Cambridge, UK; Department of Haematology, University of Cambridge, CB2 0AW Cambridge, UK
| | - Juerg Schwaller
- University Children's Hospital and Department of Biomedicine (DBM), University of Basel, 4031 Basel, Switzerland.
| | - Simón Méndez-Ferrer
- Wellcome-MRC Cambridge Stem Cell Institute, CB2 0AW Cambridge, UK; Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.
| |
Collapse
|