1
|
Safi R, Mohsen-Kanson T, Kouzi F, El-Saghir J, Dermesrobian V, Zugasti I, Zibara K, Menéndez P, El Hajj H, El-Sabban M. Direct Interaction Between CD34 + Hematopoietic Stem Cells and Mesenchymal Stem Cells Reciprocally Preserves Stemness. Cancers (Basel) 2024; 16:3972. [PMID: 39682159 DOI: 10.3390/cancers16233972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES A specialized microenvironment in the bone marrow, composed of stromal cells including mesenchymal stem cells (MSCs), supports hematopoietic stem cell (HSC) self-renewal, and differentiation bands play an important role in leukemia development and progression. The reciprocal direct interaction between MSCs and CD34+ HSCs under physiological and pathological conditions is yet to be fully characterized. METHODS Here, we established a direct co-culture model between MSCs and CD34+ HSCs or MSCs and acute myeloid leukemia cells (THP-1, Molm-13, and primary cells from patients) to study heterocellular communication. RESULTS Following MSCs-CD34+ HSCs co-culture, the expression of adhesion markers N-Cadherin and connexin 43 increased in both cell types, forming gap junction channels. Moreover, the clonogenic potential of CD34+ HSCs was increased. However, direct contact of acute myeloid leukemia cells with MSCs reduced the expression levels of connexin 43 and N-Cadherin in MSCs. The impairment in gap junction formation may potentially be due to a defect in the acute myeloid leukemia-derived MSCs. Interestingly, CD34+ HSCs and acute myeloid leukemia cell lines attenuated MSC osteoblastic differentiation upon prolonged direct cell-cell contact. CONCLUSIONS In conclusion, under physiological conditions, connexin 43 and N-Cadherin interaction preserves stemness of both CD34+ HSCs and MSCs, a process that is compromised in acute myeloid leukemia, pointing to the possible role of gap junctions in modulating stemness.
Collapse
Affiliation(s)
- Rémi Safi
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
- Josep Carreras Leukemia Research Institute, 08916 Barcelona, Spain
| | - Tala Mohsen-Kanson
- Faculty of Science, Lebanese University, Zahle 1801, Lebanon
- Faculty of Science, Lebanese University, Hadath 40016, Lebanon
| | - Farah Kouzi
- Faculty of Science, Lebanese University, Zahle 1801, Lebanon
- Faculty of Science, Lebanese University, Hadath 40016, Lebanon
| | - Jamal El-Saghir
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vera Dermesrobian
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
- Laboratory of Adaptive Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Inés Zugasti
- Department of Hematology, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - Kazem Zibara
- Faculty of Science, Lebanese University, Zahle 1801, Lebanon
- Faculty of Science, Lebanese University, Hadath 40016, Lebanon
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, 08916 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
- Consorcio Investigación Biomédica en Red de Cancer, CIBER-ONC, ISCIII, 28029 Barcelona, Spain
- Spanish Network for Advanced Cell Therapies (TERAV), 08028 Barcelona, Spain
| | - Hiba El Hajj
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
| |
Collapse
|
2
|
Moghassemi S, Dadashzadeh A, Lucci CM, Amorim CA. Tumor-Infiltration Mimicking Model of Contaminated Ovarian Tissue as an Innovative Platform for Advanced Cancer Research. AAPS J 2024; 27:7. [PMID: 39586867 DOI: 10.1208/s12248-024-00997-5] [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: 09/10/2024] [Accepted: 11/09/2024] [Indexed: 11/27/2024] Open
Abstract
The development of advanced preclinical models is crucial for the evaluation and validation of novel therapeutic strategies in oncology. Three-dimensional (3D) microtumor models, which incorporate both cancer and stromal cells within biomimetic hydrogels, have emerged as powerful tools that more accurately replicate the complex tumor microenvironment compared to traditional two-dimensional (2D) cell culture systems. In this context, our study aims to develop 3D microtumor models by integrating cancer and stromal cells within an extracellular-matrix-mimetic hydrogel, as a physiologically accurate microtumor model that can serve as an innovative platform for advanced cancer research and drug screening. Microtumors composed of varying ratios of leukemia cells (HL-60) to healthy ovarian stromal cells (SCs) (1:1, 1:10, 1:100, or 1:1000) were encapsulated in PEGylated fibrin hydrogel and cultured for 5 days. The proliferation and dynamics of cancerous and healthy cell populations were evaluated using CD43/Ki67 immunofluorescence double staining. Our findings indicate that tumor development and malignancy progression can be influenced by adjusting cell culture ratios and incubation time. Notably, the HL-60:SCs ratio of 1:100 closely replicated leukemia cell invasion in ovarian tissue, demonstrating detectable malignancy on the third and fifth days without significant changes in total cell density dynamics. This 3D leukemia microtumor model offers superior physiological relevance compared to traditional 2D in vitro assays and shows promising potential for applications in cellular analysis and drug screening.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Hippocrate 54, Bte B1.55.03, 1200, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Hippocrate 54, Bte B1.55.03, 1200, Brussels, Belgium
| | - Carolina M Lucci
- Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Christiani A Amorim
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Hippocrate 54, Bte B1.55.03, 1200, Brussels, Belgium.
| |
Collapse
|
3
|
Wilczyński B, Dąbrowska A, Kulbacka J, Baczyńska D. Chemoresistance and the tumor microenvironment: the critical role of cell-cell communication. Cell Commun Signal 2024; 22:486. [PMID: 39390572 PMCID: PMC11468187 DOI: 10.1186/s12964-024-01857-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024] Open
Abstract
Resistance of cancer cells to anticancer drugs remains a major challenge in modern medicine. Understanding the mechanisms behind the development of chemoresistance is key to developing appropriate therapies to counteract it. Nowadays, with advances in technology, we are paying more and more attention to the role of the tumor microenvironment (TME) and intercellular interactions in this process. We also know that important elements of the TME are not only the tumor cells themselves but also other cell types, such as mesenchymal stem cells, cancer-associated fibroblasts, stromal cells, and macrophages. TME elements can communicate with each other indirectly (via cytokines, chemokines, growth factors, and extracellular vesicles [EVs]) and directly (via gap junctions, ligand-receptor pairs, cell adhesion, and tunnel nanotubes). This communication appears to be critical for the development of chemoresistance. EVs seem to be particularly interesting structures in this regard. Within these structures, lipids, proteins, and nucleic acids can be transported, acting as signaling molecules that interact with numerous biochemical pathways, thereby contributing to chemoresistance. Moreover, drug efflux pumps, which are responsible for removing drugs from cancer cells, can also be transported via EVs.
Collapse
Affiliation(s)
- Bartosz Wilczyński
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, Wroclaw, 50-367, Poland
| | - Alicja Dąbrowska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, Wroclaw, 50-367, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, Wroclaw, 50-556, Poland.
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Santariškių g. 5, Vilnius, LT-08406, Lithuania.
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, Wroclaw, 50-556, Poland
| |
Collapse
|
4
|
Edwards E, Schenone D, Sivagnanalingam U, Perry S, Mullen CA. GAP JUNCTION FUNCTION IS ESSENTIAL FOR SURVIVAL OF ACUTE LYMPHOBLASTIC LEUKEMIA CELLS. Exp Oncol 2024; 46:110-118. [PMID: 39396173 DOI: 10.15407/exp-oncology.2024.02.110] [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: 10/09/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Acute lymphoblastic leukemia has an intimate physical relationship with nonmalignant bone marrow stromal cells. We have recently demonstrated that stromal cells contribute to the survival of leukemia cells and that there is a bidirectional transfer of intracellular material between them. Understanding the mechanisms of stromal support of leukemia may provide insights into new therapies. AIM To test the hypothesis that gap junctions are formed between acute lymphoblastic leukemia cells and nonmalignant stromal cells, and that gap junction function is essential for the survival of leukemia cells. MATERIALS AND METHODS We employed a well-characterized in vitro model of human bone marrow stromal cells and primary human B lymphoblastic leukemia cells and measured leukemia cell survival in coculture using flow cytometry. We measured the effects of gap junction antagonist peptides, carbenoxolone (a drug known to interfere with the gap junction function), and several leukemia chemotherapy drugs including methotrexate upon leukemia cell survival. RESULTS We demonstrated that stromal cells need to be alive and metabolically active to keep leukemia cells alive. Physical contact between stromal and leukemia cells leads to an increase in gap junction proteins in leukemia cells. Gap junction inhibitory peptides impaired leukemia cell survival as did carbenoxolone, a nonpeptide inhibitor of the gap junction function. Stromal cell survival was not affected. We observed a very modest enhancement of methotrexate antileukemia activity by low-dose carbenoxolone but no significant interactions with dexamethasone, vincristine, mercaptopurine, or doxorubicin. CONCLUSION These studies demonstrate that acute lymphoblastic cell survival is impaired by interference with the gap junction function. The development of drugs targeting gap junctions may provide a novel approach to the therapy of acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- E Edwards
- Division of Pediatric Hematology/Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - D Schenone
- Division of Pediatric Hematology/Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - U Sivagnanalingam
- Division of Pediatric Hematology/Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - S Perry
- Division of Pediatric Hematology/Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - C A Mullen
- Division of Pediatric Hematology/Oncology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
5
|
Li D, Shao F, Yu Q, Wu R, Tuo Z, Wang J, Ye L, Guo Y, Yoo KH, Ke M, Okoli UA, Premkamon C, Yang Y, Wei W, Heavey S, Cho WC, Feng D. The complex interplay of tumor-infiltrating cells in driving therapeutic resistance pathways. Cell Commun Signal 2024; 22:405. [PMID: 39160622 PMCID: PMC11331645 DOI: 10.1186/s12964-024-01776-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024] Open
Abstract
Drug resistance remains a significant challenge in cancer treatment. Recently, the interactions among various cell types within the tumor microenvironment (TME) have deepened our understanding of the mechanisms behind treatment resistance. Therefore, this review aims to synthesize current research focusing on infiltrating cells and drug resistance suggesting that targeting the TME could be a viable strategy to combat this issue. Numerous factors, including inflammation, metabolism, senescence, hypoxia, and angiogenesis, contribute to drug resistance could be a viable strategy to combat this issue. Overexpression of STAT3 is commonly associated with drug-resistant cancer cells or stromal cells. Current research often generalizes the impact of stromal cells on resistance, lacking specificity and statistical robustness. Thus, future research should take notice of this issue and aim to provide high-quality evidence. Despite the existing limitations, targeting the TME to overcome therapy resistance hold promising and valuable potential.
Collapse
Affiliation(s)
- Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fanglin Shao
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qingxin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, 315211, China
- Department of Pathology, Ningbo Medical Centre Lihuili Hospital, Ningbo, China
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Yiqing Guo
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, Seoul, Republic of Korea
| | - Mang Ke
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Uzoamaka Adaobi Okoli
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK
- Basic and Translational Cancer Research Group, Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria, Eastern part of Nigeria, Nsukka, Enugu State, Nigeria
| | - Chaipanichkul Premkamon
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK
| | - Yubo Yang
- Department of Urology, Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, 404000, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Susan Heavey
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Birmingham, Hong Kong SAR, China.
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK.
| |
Collapse
|
6
|
Urs AP, Goda C, Kulkarni R. Remodeling of the bone marrow microenvironment during acute myeloid leukemia progression. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:63. [PMID: 39118939 PMCID: PMC11304419 DOI: 10.21037/atm-23-1824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/23/2023] [Indexed: 08/10/2024]
Abstract
Hematopoiesis requires a complex interplay between the hematopoietic stem and progenitor cells and the cells of the bone marrow microenvironment (BMM). The BMM is heterogeneous, with different regions having distinct cellular, molecular, and metabolic composition and function. Studies have shown that this niche is disrupted in patients with acute myeloid leukemia (AML), which plays a crucial role in disease progression. This review provides a comprehensive overview of the components of vascular and endosteal niches and the molecular mechanisms by which they regulate normal hematopoiesis. We also discuss how these niches are modified in the context of AML, into a disease-promoting niche and how the modified niches in turn regulate AML blast survival and proliferation. We focus on mechanisms of modifications in structural and cellular components of the bone marrow (BM) niche by the AML cells and its impact on leukemic progression and patient outcome. Finally, we also discuss mechanisms by which the altered BM niche protects AML blasts from treatment agents, thereby causing therapy resistance in AML patients. We also summarize ongoing clinical trials that target various BM niche components in the treatment of AML patients. Hence, the BM niche represents a promising target to treat AML and promote normal hematopoiesis.
Collapse
Affiliation(s)
- Amog P. Urs
- The Division of Hematology and Hematological Malignancies, Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT, USA
| | - Chinmayee Goda
- The Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Rohan Kulkarni
- The Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
7
|
Kou R, Li T, Fu C, Jiang D, Wang Y, Meng J, Zhong R, Liang C, Dong M. Exosome-shuttled FTO from BM-MSCs contributes to cancer malignancy and chemoresistance in acute myeloid leukemia by inducing m6A-demethylation: A nano-based investigation. ENVIRONMENTAL RESEARCH 2024; 244:117783. [PMID: 38048862 DOI: 10.1016/j.envres.2023.117783] [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: 05/31/2023] [Revised: 11/09/2023] [Accepted: 11/23/2023] [Indexed: 12/06/2023]
Abstract
Although bone marrow mesenchymal stem cells (BM-MSCs)-derived exosomes have been reported to be closely associated with acute myeloid leukemia (AML) progression and chemo-resistance, but its detailed functions and molecular mechanisms have not been fully delineated. Besides, serum RNA m6A demethylase fat mass and obesity-associated protein (FTO)-containing exosomes are deemed as important indicators for cancer progression, and this study aimed to investigate the role of BM-MSCs-derived FTO-exosomes in regulating the malignant phenotypes of AML cells. Here, we verified that BM-MSCs-derived exosomes delivered FTO to promote cancer aggressiveness, stem cell properties and Cytosine arabinoside (Ara-C)-chemoresistance in AML cells, and the underlying mechanisms were also uncovered. Our data suggested that BM-MSCs-derived FTO-exo demethylated m6A modifications in the m6A-modified LncRNA GLCC1 to facilitate its combination with the RNA-binding protein Hu antigen R (HuR), which further increased the stability and expression levels of LncRNA GLCC1. In addition, LncRNA GLCC1 was verified as an oncogene to facilitate cell proliferation and enhanced Ara-C-chemoresistance in AML cells. Further experiments confirmed that demethylated LncRNA GLCC1 served as scaffold to facilitate the formation of the IGF2 mRNA binding protein 1 (IGF2BP1)-c-Myc complex, which led to the activation of the downstream tumor-promoting c-Myc-associated signal pathways. Moreover, our rescuing experiments validated that the promoting effects of BM-MSCs-derived FTO-exo on cancer aggressiveness and drug resistance in AML cells were abrogated by silencing LncRNA GLCC1 and c-Myc. Thus, the present firstly investigated the functions and underlying mechanisms by which BM-MSCs-derived FTO-exo enhanced cancer aggressiveness and chemo-resistance in AML by modulating the LncRNA GLCC1-IGF2BP1-c-Myc signal pathway, and our work provided novel biomarkers for the diagnosis, treatment and therapy of AML in clinic.
Collapse
Affiliation(s)
- Ruirui Kou
- Department of Hematology, The Second Affiliated Hospital of Hainan Medical College, Yehai Road No. 368, Longhua District, Haikou, 570000, Hainan Province, China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Caizhu Fu
- Department of Hematology, The Second Affiliated Hospital of Hainan Medical College, Yehai Road No. 368, Longhua District, Haikou, 570000, Hainan Province, China.
| | - Duanfeng Jiang
- Department of Hematology, The Second Affiliated Hospital of Hainan Medical College, Yehai Road No. 368, Longhua District, Haikou, 570000, Hainan Province, China.
| | - Yue Wang
- Department of Pharmacology and Toxicology, Wright State University, USA.
| | - Jie Meng
- Department of Hematology, The Second Affiliated Hospital of Hainan Medical College, Yehai Road No. 368, Longhua District, Haikou, 570000, Hainan Province, China.
| | - Ruilan Zhong
- Department of Hematology, The Second Affiliated Hospital of Hainan Medical College, Yehai Road No. 368, Longhua District, Haikou, 570000, Hainan Province, China.
| | - Changjiu Liang
- Department of Hematology, The Second Affiliated Hospital of Hainan Medical College, Yehai Road No. 368, Longhua District, Haikou, 570000, Hainan Province, China.
| | - Min Dong
- Department of Hematology, The Second Affiliated Hospital of Hainan Medical College, Yehai Road No. 368, Longhua District, Haikou, 570000, Hainan Province, China.
| |
Collapse
|
8
|
Liesveld J, Galipeau J. In Vitro Insights Into the Influence of Marrow Mesodermal/Mesenchymal Progenitor Cells on Acute Myelogenous Leukemia and Myelodysplastic Syndromes. Stem Cells 2023; 41:823-836. [PMID: 37348128 DOI: 10.1093/stmcls/sxad050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023]
Abstract
The study of marrow-resident mesodermal progenitors can provide important insight into their role in influencing normal and aberrant hematopoiesis as occurs in acute myelogenous leukemia (AML) and myelodysplastic syndromes (MDS). In addition, the chemokine competency of these cells provides links to the inflammatory milieu of the marrow microenvironment with additional implications for normal and malignant hematopoiesis. While in vivo studies have elucidated the structure and function of the marrow niche in murine genetic models, corollary human studies have not been feasible, and thus the use of culture-adapted mesodermal cells has provided insights into the role these rare endogenous niche cells play in physiologic, malignant, and inflammatory states. This review focuses on culture-adapted human mesenchymal stem/stromal cells (MSCs) as they have been utilized in understanding their influence in AML and MDS as well as on their chemokine-mediated responses to myeloid malignancies, injury, and inflammation. Such studies have intrinsic limitations but have provided mechanistic insights and clues regarding novel druggable targets.
Collapse
Affiliation(s)
- Jane Liesveld
- Department of Medicine, James P. Wilmot Cancer Institute, University of Rochester, Rochester, NY, USA
| | - Jaques Galipeau
- University of Wisconsin School of Medicine and Public Health, University of Wisconsin in Madison, Madison, WI, USA
| |
Collapse
|
9
|
Feng L, Zhang PY, Gao W, Yu J, Robson SC. Targeting chemoresistance and mitochondria-dependent metabolic reprogramming in acute myeloid leukemia. Front Oncol 2023; 13:1244280. [PMID: 37746249 PMCID: PMC10513429 DOI: 10.3389/fonc.2023.1244280] [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: 06/22/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Chemoresistance often complicates the management of cancer, as noted in the instance of acute myeloid leukemia (AML). Mitochondrial function is considered important for the viability of AML blasts and appears to also modulate chemoresistance. As mitochondrial metabolism is aberrant in AML, any distinct pathways could be directly targeted to impact both cell viability and chemoresistance. Therefore, identifying and targeting those precise rogue elements of mitochondrial metabolism could be a valid therapeutic strategy in leukemia. Here, we review the evidence for abnormalities in mitochondria metabolic processes in AML cells, that likely impact chemoresistance. We further address several therapeutic approaches targeting isocitrate dehydrogenase 2 (IDH2), CD39, nicotinamide phosphoribosyl transferase (NAMPT), electron transport chain (ETC) complex in AML and also consider the roles of mesenchymal stromal cells. We propose the term "mitotherapy" to collectively refer to such regimens that attempt to override mitochondria-mediated metabolic reprogramming, as used by cancer cells. Mounting evidence suggests that mitotherapy could provide a complementary strategy to overcome chemoresistance in liquid cancers, as well as in solid tumors.
Collapse
Affiliation(s)
- Lili Feng
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Philip Y. Zhang
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Wenda Gao
- Antagen Institute for Biomedical Research, Canton, MA, United States
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Simon C. Robson
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Department of Medicine, Division of Gastroenterology/Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
10
|
Chai C, Sui K, Tang J, Yu H, Yang C, Zhang H, Li SC, Zhong JF, Wang Z, Zhang X. BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications imply therapies of leukemic subclonal evolution. Theranostics 2023; 13:3943-3963. [PMID: 37554265 PMCID: PMC10405834 DOI: 10.7150/thno.83178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/12/2023] [Indexed: 08/10/2023] Open
Abstract
Rationale: In the bone marrow microenvironment (BMME), mesenchymal stem/stromal cells (MSCs) control the self-renewal of both healthy and cancerous hematopoietic stem/progenitor cells (HSPCs). We previously showed that in vivo leukemia-derived MSCs change neighbor MSCs into leukemia-permissive states and boost leukemia cell proliferation, survival, and chemotherapy resistance. But the mechanisms behind how the state changes are still not fully understood. Methods: Here, we took a reverse engineering approach to determine BCR-ABL1+ leukemia cells activated transcriptional factor C/EBPβ, resulting in miR130a/b-3p production. Then, we back-tracked from clinical specimen transcriptome sequencing to cell co-culture, molecular and cellular assays, flow cytometry, single-cell transcriptome, and transcriptional regulation to determine the molecular mechanisms of BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications. Results: BCR-ABL1-driven exosome-miR130a/b-3p mediated gap-junction Cx43 (a.k.a., GJA1) BMSC intercellular communications for subclonal evolution in leukemic microenvironment by targeting BMSCs-expressed HLAs, thereby potentially maintaining BMSCs with self-renewal properties and reduced BMSC immunogenicity. The Cx43low and miR-130a/bhigh subclonal MSCs subsets of differentiation state could be reversed to Cx43high and miR-130a/blow subclones of the higher stemness state in Cx43-overexpressed subclonal MSCs. Both miR-130a and miR-130b might only inhibit Cx43 translation or degrade Cx43 proteins and did not affect Cx43 mRNA stability. The subclonal evolution was further confirmed by single-cell transcriptome profiling of MSCs, which suggested that Cx43 regulated their stemness and played normal roles in immunomodulation antigen processing. Thus, upregulated miR-130a/b promoted osteogenesis and adipogenesis from BMSCs, thereby decreasing cancer progression. Our clinical data validated that the expression of many genes in human major histocompatibility was negatively associated with the stemness of MSCs, and several immune checkpoint proteins contributing to immune escape in tumors were overexpressed after either miR-130a or miR-130b overexpression, such as CD274, LAG3, PDCD1, and TNFRSF4. Not only did immune response-related cytokine-cytokine receptor interactions and PI3K-AKT pathways, including EGR3, TNFRSF1B, but also NDRG2 leukemic-associated inflammatory factors, such as IFNB1, CXCL1, CXCL10, and CCL7 manifest upon miR-130a/b overexpression. Either BCR siRNAs or ABL1 siRNAs assay showed significantly decreased miR-130a and miR-130b expression, and chromatin immunoprecipitation sequencing confirmed that the regulation of miR-130a and miR-130b expression is BCR-ABL1-dependent. BCR-ABL1 induces miR-130a/b expression through the upregulation of transcriptional factor C/EBPβ. C/EBPβ could bind directly to the promoter region of miR-130b-3p, not miR-130a-3p. BCR-ABL1-driven exosome-miR130a-3p could interact with Cx43, and further impact GJIC in TME. Conclusion: Our findings shed light on how leukemia BCR-ABL1-driven exosome-miR130b-3p could interact with gap-junction Cx43, and further impact GJIC in TME, implications for leukemic therapies of subclonal evolution.
Collapse
Affiliation(s)
- Chengyan Chai
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Ke Sui
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Jun Tang
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
| | - Hao Yu
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Chao Yang
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Hongyang Zhang
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
| | - Shengwen Calvin Li
- Neuro-Oncology and Stem Cell Research Laboratory, Center for Neuroscience Research, CHOC Children's Research Institute, Children's Hospital of Orange County (CHOC), 1201 La Veta Ave., Orange, CA 92868-3874, United States of America
- Department of Neurology, University of California-Irvine School of Medicine, 200 S. Manchester Ave. Ste. 206, Orange, CA 92868, United States of America
| | - Jiang F. Zhong
- Department of Basic Sciences, Loma Linda University, Loma Linda, California, 92354, United States of America
| | - Zheng Wang
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Xi Zhang
- Medical Center of Hematology, Second Affiliated Hospital, Army Medical University, Chongqing,400037, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| |
Collapse
|
11
|
Lehner KM, Gopalakrishnapillai A, Kolb EA, Barwe SP. Bone Marrow Microenvironment-Induced Chemoprotection in KMT2A Rearranged Pediatric AML Is Overcome by Azacitidine-Panobinostat Combination. Cancers (Basel) 2023; 15:3112. [PMID: 37370721 DOI: 10.3390/cancers15123112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Advances in therapies of pediatric acute myeloid leukemia (AML) have been minimal in recent decades. Although 82% of patients will have an initial remission after intensive therapy, approximately 40% will relapse. KMT2A is the most common chromosomal translocation in AML and has a poor prognosis resulting in high relapse rates and low chemotherapy efficacy. Novel targeted approaches are needed to increase sensitivity to chemotherapy. Recent studies have shown how interactions within the bone marrow (BM) microenvironment help AML cells evade chemotherapy and contribute to relapse by promoting leukemic blast survival. This study investigates how DNA hypomethylating agent azacitidine and histone deacetylase inhibitor panobinostat synergistically overcome BM niche-induced chemoprotection modulated by stromal, endothelial, and mesenchymal stem cells and the extracellular matrix (ECM). We show that direct contact between AML cells and BM components mediates chemoprotection. We demonstrate that azacitidine and panobinostat synergistically sensitize MV4;11 cells and KMT2A rearranged pediatric patient-derived xenograft lines to cytarabine in multicell coculture. Treatment with the epigenetic drug combination reduced leukemic cell association with multicell monolayer and ECM in vitro and increased mobilization of leukemic cells from the BM in vivo. Finally, we show that pretreatment with the epigenetic drug combination improves the efficacy of chemotherapy in vivo.
Collapse
Affiliation(s)
- Kara M Lehner
- Lisa Dean Moseley Foundation Institute for Cancer and Blood Disorders, Nemours Children's Hospital, Wilmington, DE 19803, USA
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Anilkumar Gopalakrishnapillai
- Lisa Dean Moseley Foundation Institute for Cancer and Blood Disorders, Nemours Children's Hospital, Wilmington, DE 19803, USA
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Edward Anders Kolb
- Lisa Dean Moseley Foundation Institute for Cancer and Blood Disorders, Nemours Children's Hospital, Wilmington, DE 19803, USA
| | - Sonali P Barwe
- Lisa Dean Moseley Foundation Institute for Cancer and Blood Disorders, Nemours Children's Hospital, Wilmington, DE 19803, USA
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
12
|
Xing J, Wang R, Cui F, Song L, Ma Q, Xu H. Role of the regulation of mesenchymal stem cells on macrophages in sepsis. Int J Immunopathol Pharmacol 2023; 37:3946320221150722. [PMID: 36840553 PMCID: PMC9969469 DOI: 10.1177/03946320221150722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
Sepsis is a common clinical critical disease with high mortality. The excessive release of cytokines from macrophages is the main cause of out-of-control immune response in sepsis. Mesenchymal stem cells (MSCs) are thought to be useful in adjunctive therapy of sepsis and related diseases, due to their function in immune regulation, anti-inflammatory, antibacterial, and tissue regeneration. Also there have been several successful cases in clinical treatment. Some previous studies have shown that MSCs regulate the function of macrophages through secreting cytokines and extracellular vesicles, or transferring mitochondria directly to target cells, which affects the progress of sepsis. Here, we review the regulation of MSCs on macrophages in sepsis, mainly focus on the regulation ways. We hope that will help to understand the immunological mechanism and also provide some clues for the clinical application of MSCs in the biotherapy of sepsis.
Collapse
Affiliation(s)
- Jie Xing
- Fenyang Hospital of Shanxi
Province, Fenyang, China
| | - Rui Wang
- School of Life Sciences, Northwestern Polytechnical
University, Xi’an, China
| | - Fengqi Cui
- School of Life Sciences, Northwestern Polytechnical
University, Xi’an, China
| | - Linlin Song
- Fenyang Hospital of Shanxi
Province, Fenyang, China
| | - Quanlin Ma
- Fenyang Hospital of Shanxi
Province, Fenyang, China,Quanlin Ma, Department of Cardiothoracic
Surgery, Fenyang Hospital of Shanxi Province, 186 Shengli Street, Fenyang
032200, China.
| | - Huiyun Xu
- School of Life Sciences, Northwestern Polytechnical
University, Xi’an, China,Huiyun Xu, School of Life Sciences,
Northwestern Polytechnical University, 127 Youyi Xilu, Xi’an 710072, China.
| |
Collapse
|
13
|
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
|
14
|
Chen F, Zhong X, Dai Q, Li K, Zhang W, Wang J, Zhao Y, Shen J, Xiao Z, Xing H, Li J. Human Umbilical Cord MSC Delivered-Soluble TRAIL Inhibits the Proliferation and Promotes Apoptosis of B-ALL Cell In Vitro and In Vivo. Pharmaceuticals (Basel) 2022; 15:1391. [PMID: 36422522 PMCID: PMC9693801 DOI: 10.3390/ph15111391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 09/19/2023] Open
Abstract
The TNF-related apoptosis-inducing ligand (TRAIL) could induce apoptosis of leukemic cells, while showed no cytotoxic effect on normal cells. One of the limitations for application of recombinant TRAIL (rhTRAIL) in leukemia treatment is that the serum half-life of this protein is short. Gene delivery is a good strategy to prolong the half-life of TRAIL. In this study, we genetically engineered umbilical cord-MSCs to continuously express and secrete soluble TRAIL (MSC-sTRAIL), to investigate the effects of MSC-sTRAIL on B-cell acute lymphocytic leukemia (B-ALL) cells. In vitro, MSC-sTRAIL significantly inhibited the proliferation of B-ALL cells by suppressing PI3K/AKT and MEK/ERK signaling pathways, and induced apoptosis of B-ALL cells via the caspase cascade-mediated pathway and mitochondrial-mediated pathway. In vivo, MSC-sTRAIL dramatically inhibited B-ALL cell growth. Meanwhile, B-ALL-induced splenic and renal injuries were significantly alleviated after MSC-sTRAIL treatment. Moreover, the serum levels of MSC-secreted sTRAIL were still high in MSC-sTRAIL treated mice, indicating an extended half-life of sTRAIL. Our study suggests that MSC delivered-TRAIL secretion is a potential therapeutic strategy for B-ALL treatment.
Collapse
Affiliation(s)
- Fangshan Chen
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xianmei Zhong
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
- Department of Pharmacy, People’s Hospital of Nanbu County, Nanchong 637300, China
| | - Qian Dai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Kuo Li
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Wei Zhang
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jie Wang
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Hongyun Xing
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jing Li
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
15
|
Vilaplana-Lopera N, Cuminetti V, Almaghrabi R, Papatzikas G, Rout AK, Jeeves M, González E, Alyahyawi Y, Cunningham A, Erdem A, Schnütgen F, Raghavan M, Potluri S, Cazier JB, Schuringa JJ, Reed MAC, Arranz L, Günther UL, Garcia P. Crosstalk between AML and stromal cells triggers acetate secretion through the metabolic rewiring of stromal cells. eLife 2022; 11:e75908. [PMID: 36052997 PMCID: PMC9477493 DOI: 10.7554/elife.75908] [Citation(s) in RCA: 10] [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: 11/26/2021] [Accepted: 09/01/2022] [Indexed: 11/18/2022] Open
Abstract
Acute myeloid leukaemia (AML) cells interact and modulate components of their surrounding microenvironment into their own benefit. Stromal cells have been shown to support AML survival and progression through various mechanisms. Nonetheless, whether AML cells could establish beneficial metabolic interactions with stromal cells is underexplored. By using a combination of human AML cell lines and AML patient samples together with mouse stromal cells and a MLL-AF9 mouse model, here we identify a novel metabolic crosstalk between AML and stromal cells where AML cells prompt stromal cells to secrete acetate for their own consumption to feed the tricarboxylic acid cycle (TCA) and lipid biosynthesis. By performing transcriptome analysis and tracer-based metabolic NMR analysis, we observe that stromal cells present a higher rate of glycolysis when co-cultured with AML cells. We also find that acetate in stromal cells is derived from pyruvate via chemical conversion under the influence of reactive oxygen species (ROS) following ROS transfer from AML to stromal cells via gap junctions. Overall, we present a unique metabolic communication between AML and stromal cells and propose two different molecular targets, ACSS2 and gap junctions, that could potentially be exploited for adjuvant therapy.
Collapse
Affiliation(s)
- Nuria Vilaplana-Lopera
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| | - Vincent Cuminetti
- Stem Cells, Ageing and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT – The Arctic University of NorwayTromsoNorway
| | - Ruba Almaghrabi
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Department of Laboratory Medicine (hematology), Faculty of Applied Medical Sciences. Albaha University, Kingdom of Saudi ArabiaAl BahahSaudi Arabia
| | - Grigorios Papatzikas
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Centre for Computational Biology, University of BirminghamBirminghamUnited Kingdom
| | - Ashok Kumar Rout
- Institute of Chemistry and Metabolomics, University of LübeckLübeckGermany
| | - Mark Jeeves
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| | - Elena González
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| | - Yara Alyahyawi
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan UniversityJazanSaudi Arabia
| | - Alan Cunningham
- Department of Experimental Hematology, University Medical Center Groningen, University of GroningenGroningenNetherlands
| | - Ayşegül Erdem
- Department of Experimental Hematology, University Medical Center Groningen, University of GroningenGroningenNetherlands
| | - Frank Schnütgen
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University FrankfurtFrankfurtGermany
- Frankfurt Cancer Institute, Goethe University FrankfurtFrankfurtGermany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, and German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Manoj Raghavan
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical CentreBirminghamUnited Kingdom
| | - Sandeep Potluri
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical CentreBirminghamUnited Kingdom
| | - Jean-Baptiste Cazier
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Centre for Computational Biology, University of BirminghamBirminghamUnited Kingdom
| | - Jan Jacob Schuringa
- Department of Experimental Hematology, University Medical Center Groningen, University of GroningenGroningenNetherlands
| | - Michelle AC Reed
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| | - Lorena Arranz
- Stem Cells, Ageing and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT – The Arctic University of NorwayTromsoNorway
| | - Ulrich L Günther
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
- Institute of Chemistry and Metabolomics, University of LübeckLübeckGermany
| | - Paloma Garcia
- Institute of Cancer and Genomic Sciences, University of BirminghamBirminghamUnited Kingdom
| |
Collapse
|
16
|
Properties of Leukemic Stem Cells in Regulating Drug Resistance in Acute and Chronic Myeloid Leukemias. Biomedicines 2022; 10:biomedicines10081841. [PMID: 36009388 PMCID: PMC9405586 DOI: 10.3390/biomedicines10081841] [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: 07/04/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Notoriously known for their capacity to reconstitute hematological malignancies in vivo, leukemic stem cells (LSCs) represent key drivers of therapeutic resistance and disease relapse, posing as a major medical dilemma. Despite having low abundance in the bulk leukemic population, LSCs have developed unique molecular dependencies and intricate signaling networks to enable self-renewal, quiescence, and drug resistance. To illustrate the multi-dimensional landscape of LSC-mediated leukemogenesis, in this review, we present phenotypical characteristics of LSCs, address the LSC-associated leukemic stromal microenvironment, highlight molecular aberrations that occur in the transcriptome, epigenome, proteome, and metabolome of LSCs, and showcase promising novel therapeutic strategies that potentially target the molecular vulnerabilities of LSCs.
Collapse
|
17
|
Salazar-Terreros MJ, Vernot JP. In Vitro and In Vivo Modeling of Normal and Leukemic Bone Marrow Niches: Cellular Senescence Contribution to Leukemia Induction and Progression. Int J Mol Sci 2022; 23:7350. [PMID: 35806354 PMCID: PMC9266537 DOI: 10.3390/ijms23137350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 12/16/2022] Open
Abstract
Cellular senescence is recognized as a dynamic process in which cells evolve and adapt in a context dependent manner; consequently, senescent cells can exert both beneficial and deleterious effects on their surroundings. Specifically, senescent mesenchymal stromal cells (MSC) in the bone marrow (BM) have been linked to the generation of a supporting microenvironment that enhances malignant cell survival. However, the study of MSC's senescence role in leukemia development has been straitened not only by the availability of suitable models that faithfully reflect the structural complexity and biological diversity of the events triggered in the BM, but also by the lack of a universal, standardized method to measure senescence. Despite these constraints, two- and three dimensional in vitro models have been continuously improved in terms of cell culture techniques, support materials and analysis methods; in addition, research on animal models tends to focus on the development of techniques that allow tracking leukemic and senescent cells in the living organism, as well as to modify the available mice strains to generate individuals that mimic human BM characteristics. Here, we present the main advances in leukemic niche modeling, discussing advantages and limitations of the different systems, focusing on the contribution of senescent MSC to leukemia progression.
Collapse
Affiliation(s)
- Myriam Janeth Salazar-Terreros
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogota 111321, Colombia;
| | - Jean-Paul Vernot
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogota 111321, Colombia;
- Instituto de Investigaciones Biomédicas, Facultad de Medicina, Universidad Nacional de Colombia, Bogota 111321, Colombia
| |
Collapse
|
18
|
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
|
19
|
Dakik H, El Dor M, Bourgeais J, Kouzi F, Herault O, Gouilleux F, Zibara K, Mazurier F. Diphenyleneiodonium Triggers Cell Death of Acute Myeloid Leukemia Cells by Blocking the Mitochondrial Respiratory Chain, and Synergizes with Cytarabine. Cancers (Basel) 2022; 14:cancers14102485. [PMID: 35626090 PMCID: PMC9140039 DOI: 10.3390/cancers14102485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Acute myeloid leukemia (AML) is an aggressive heterogeneous cancer of the blood, of which 70% of cases develop relapse. Relapse is mainly due to chemoresistant leukemic cells (LCs) that are characterized by high mitochondrial oxidative phosphorylation (OxPhos) status, i.e., cells that are dependent on the mitochondrial respiratory chain (MRC) function. The aim of our study was to determine whether diphenyleneiodonium (DPI)—known as a potent inhibitor of flavoproteins—could be used to target AML cells. Herein, we demonstrate that DPI disrupts the mitochondrial function of AML cell lines. Interestingly, we found that cells with high OxPhos are more sensitive to the apoptotic effects of DPI. Moreover, we showed that DPI sensitizes AML cell lines to cytarabine (Ara-C) treatment, suggesting that MRC inhibitors could be employed to target LCs that are resistant to this chemotherapeutic agent. Abstract Acute myeloid leukemia (AML) is characterized by the accumulation of undifferentiated blast cells in the bone marrow and blood. In most cases of AML, relapse frequently occurs due to resistance to chemotherapy. Compelling research results indicate that drug resistance in cancer cells is highly dependent on the intracellular levels of reactive oxygen species (ROS). Modulating ROS levels is therefore a valuable strategy to overcome the chemotherapy resistance of leukemic cells. In this study, we evaluated the efficiency of diphenyleneiodonium (DPI)—a well-known inhibitor of ROS production—in targeting AML cells. Results showed that although inhibiting cytoplasmic ROS production, DPI also triggered an increase in the mitochondrial ROS levels, caused by the disruption of the mitochondrial respiratory chain. We also demonstrated that DPI blocks mitochondrial oxidative phosphorylation (OxPhos) in a dose-dependent manner, and that AML cells with high OxPhos status are highly sensitive to treatment with DPI, which synergizes with the chemotherapeutic agent cytarabine (Ara-C). Thus, our results suggest that targeting mitochondrial function with DPI might be exploited to target AML cells with high OxPhos status.
Collapse
Affiliation(s)
- Hassan Dakik
- EA7501 GICC/CNRS ERL7001 LNOx, University of Tours, F-37032 Tours, France; (H.D.); (M.E.D.); (J.B.); (F.K.); (O.H.); (F.G.)
| | - Maya El Dor
- EA7501 GICC/CNRS ERL7001 LNOx, University of Tours, F-37032 Tours, France; (H.D.); (M.E.D.); (J.B.); (F.K.); (O.H.); (F.G.)
| | - Jérôme Bourgeais
- EA7501 GICC/CNRS ERL7001 LNOx, University of Tours, F-37032 Tours, France; (H.D.); (M.E.D.); (J.B.); (F.K.); (O.H.); (F.G.)
- Department of Biological Hematology, Tours University Hospital, F-37000 Tours, France
| | - Farah Kouzi
- EA7501 GICC/CNRS ERL7001 LNOx, University of Tours, F-37032 Tours, France; (H.D.); (M.E.D.); (J.B.); (F.K.); (O.H.); (F.G.)
- Biology Department, Faculty of Sciences, Lebanese University, Beirut 90656, Lebanon
| | - Olivier Herault
- EA7501 GICC/CNRS ERL7001 LNOx, University of Tours, F-37032 Tours, France; (H.D.); (M.E.D.); (J.B.); (F.K.); (O.H.); (F.G.)
- Department of Biological Hematology, Tours University Hospital, F-37000 Tours, France
| | - Fabrice Gouilleux
- EA7501 GICC/CNRS ERL7001 LNOx, University of Tours, F-37032 Tours, France; (H.D.); (M.E.D.); (J.B.); (F.K.); (O.H.); (F.G.)
| | - Kazem Zibara
- Biology Department, Faculty of Sciences, Lebanese University, Beirut 90656, Lebanon
- ER045, PRASE, Beirut 6573/14, Lebanon
- Correspondence: (K.Z.); (F.M.)
| | - Frédéric Mazurier
- EA7501 GICC/CNRS ERL7001 LNOx, University of Tours, F-37032 Tours, France; (H.D.); (M.E.D.); (J.B.); (F.K.); (O.H.); (F.G.)
- Correspondence: (K.Z.); (F.M.)
| |
Collapse
|
20
|
Guéguinou M, Ibrahim S, Bourgeais J, Robert A, Pathak T, Zhang X, Crottès D, Dupuy J, Ternant D, Monbet V, Guibon R, Flores-Romero H, Lefèvre A, Lerondel S, Le Pape A, Dumas JF, Frank PG, Girault A, Chautard R, Guéraud F, García-Sáez AJ, Ouaissi M, Emond P, Sire O, Hérault O, Fromont-Hankard G, Vandier C, Tougeron D, Trebak M, Raoul W, Lecomte T. Curcumin and NCLX inhibitors share anti-tumoral mechanisms in microsatellite-instability-driven colorectal cancer. Cell Mol Life Sci 2022; 79:284. [PMID: 35526196 PMCID: PMC11072810 DOI: 10.1007/s00018-022-04311-4] [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: 01/31/2022] [Revised: 04/05/2022] [Accepted: 04/15/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND AIMS Recent evidences highlight a role of the mitochondria calcium homeostasis in the development of colorectal cancer (CRC). To overcome treatment resistance, we aimed to evaluate the role of the mitochondrial sodium-calcium-lithium exchanger (NCLX) and its targeting in CRC. We also identified curcumin as a new inhibitor of NCLX. METHODS We examined whether curcumin and pharmacological compounds induced the inhibition of NCLX-mediated mitochondrial calcium (mtCa2+) extrusion, the role of redox metabolism in this process. We evaluated their anti-tumorigenic activity in vitro and in a xenograft mouse model. We analyzed NCLX expression and associations with survival in The Cancer Genome Atlas (TCGA) dataset and in tissue microarrays from 381 patients with microsatellite instability (MSI)-driven CRC. RESULTS In vitro, curcumin exerted strong anti-tumoral activity through its action on NCLX with mtCa2+ and reactive oxygen species overload associated with a mitochondrial membrane depolarization, leading to reduced ATP production and apoptosis. NCLX inhibition with pharmacological and molecular approaches reproduced the effects of curcumin. NCLX inhibitors decreased CRC tumor growth in vivo. Both transcriptomic analysis of TCGA dataset and immunohistochemical analysis of tissue microarrays demonstrated that higher NCLX expression was associated with MSI status, and for the first time, NCLX expression was significantly associated with recurrence-free survival. CONCLUSIONS Our findings highlight a novel anti-tumoral mechanism of curcumin through its action on NCLX and mitochondria calcium overload that could benefit for therapeutic schedule of patients with MSI CRC.
Collapse
Affiliation(s)
- Maxime Guéguinou
- EA 7501 GICC, Université de Tours, Tours, France.
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France.
| | | | | | - Alison Robert
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Trayambak Pathak
- Department of Cellular and Molecular Physiology, College of Medicine, The Pennsylvania State University, 500 University Dr, Hershey, PA, 17033, USA
| | - Xuexin Zhang
- Department of Cellular and Molecular Physiology, College of Medicine, The Pennsylvania State University, 500 University Dr, Hershey, PA, 17033, USA
| | - David Crottès
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Jacques Dupuy
- TOXALIM (Research Centre in Food Toxicology)-Team E9-PPCA, Université de Toulouse, UMR 1331 INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - David Ternant
- EA 7501 GICC, Université de Tours, Tours, France
- EA4245 Transplant Immunology and Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France
| | - Valérie Monbet
- IRMAR Mathematics Research Institute of Rennes, UMR-CNRS 6625, Rennes, France
| | - Roseline Guibon
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Hector Flores-Romero
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Interfaculty Institute of Biochemistry, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Antoine Lefèvre
- UMR 1253, iBrain, Université de Tours, Inserm, 37000, Tours, France
| | | | | | - Jean-François Dumas
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Philippe G Frank
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Alban Girault
- Laboratory of Cellular and Molecular Physiology, UR UPJV 4667, University of Picardie Jules Verne, Amiens, France
| | | | - Françoise Guéraud
- TOXALIM (Research Centre in Food Toxicology)-Team E9-PPCA, Université de Toulouse, UMR 1331 INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Ana J García-Sáez
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Interfaculty Institute of Biochemistry, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Mehdi Ouaissi
- EA4245 Transplant Immunology and Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France
| | - Patrick Emond
- UMR 1253, iBrain, Université de Tours, Inserm, 37000, Tours, France
| | - Olivier Sire
- IRDL Institut de Recherche Dupuy de Lôme, UMR-CNRS, 06027, Vannes, France
| | | | - Gaëlle Fromont-Hankard
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Christophe Vandier
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - David Tougeron
- Hepato-Gastroenterology Department, Poitiers University Hospital and Faculty of Medicine of Poitiers, 86000, Poitiers, France
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, College of Medicine, The Pennsylvania State University, 500 University Dr, Hershey, PA, 17033, USA
| | - William Raoul
- EA 7501 GICC, Université de Tours, Tours, France
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Thierry Lecomte
- EA 7501 GICC, Université de Tours, Tours, France.
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France.
- Department of Hepato-Gastroenterology and Digestive Oncology, CHRU de Tours, Tours, France.
| |
Collapse
|
21
|
Guillon A, Brea-Diakite D, Cezard A, Wacquiez A, Baranek T, Bourgeais J, Picou F, Vasseur V, Meyer L, Chevalier C, Auvet A, Carballido JM, Nadal Desbarats L, Dingli F, Turtoi A, Le Gouellec A, Fauvelle F, Donchet A, Crépin T, Hiemstra PS, Paget C, Loew D, Herault O, Naffakh N, Le Goffic R, Si-Tahar M. Host succinate inhibits influenza virus infection through succinylation and nuclear retention of the viral nucleoprotein. EMBO J 2022; 41:e108306. [PMID: 35506364 PMCID: PMC9194747 DOI: 10.15252/embj.2021108306] [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: 03/19/2021] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/11/2022] Open
Abstract
Influenza virus infection causes considerable morbidity and mortality, but current therapies have limited efficacy. We hypothesized that investigating the metabolic signaling during infection may help to design innovative antiviral approaches. Using bronchoalveolar lavages of infected mice, we here demonstrate that influenza virus induces a major reprogramming of lung metabolism. We focused on mitochondria‐derived succinate that accumulated both in the respiratory fluids of virus‐challenged mice and of patients with influenza pneumonia. Notably, succinate displays a potent antiviral activity in vitro as it inhibits the multiplication of influenza A/H1N1 and A/H3N2 strains and strongly decreases virus‐triggered metabolic perturbations and inflammatory responses. Moreover, mice receiving succinate intranasally showed reduced viral loads in lungs and increased survival compared to control animals. The antiviral mechanism involves a succinate‐dependent posttranslational modification, that is, succinylation, of the viral nucleoprotein at the highly conserved K87 residue. Succinylation of viral nucleoprotein altered its electrostatic interactions with viral RNA and further impaired the trafficking of viral ribonucleoprotein complexes. The finding that succinate efficiently disrupts the influenza replication cycle opens up new avenues for improved treatment of influenza pneumonia.
Collapse
Affiliation(s)
- Antoine Guillon
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France.,Service de Médecine Intensive Réanimation, CHRU de Tours, Tours, France
| | - Deborah Brea-Diakite
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Adeline Cezard
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Alan Wacquiez
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Thomas Baranek
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Jérôme Bourgeais
- Université de Tours, Tours, France.,CNRS ERL 7001 LNOx "Leukemic niche and redox metabolism", Tours, France.,Service d'Hématologie Biologique, CHRU de Tours, Tours, France
| | - Frédéric Picou
- Université de Tours, Tours, France.,CNRS ERL 7001 LNOx "Leukemic niche and redox metabolism", Tours, France.,Service d'Hématologie Biologique, CHRU de Tours, Tours, France
| | - Virginie Vasseur
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Léa Meyer
- Virologie et Immunologie Moléculaires, INRAe, Université Paris-Saclay, Jouy-en-Josas, France
| | - Christophe Chevalier
- Virologie et Immunologie Moléculaires, INRAe, Université Paris-Saclay, Jouy-en-Josas, France
| | - Adrien Auvet
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France.,Service de Médecine Intensive Réanimation, CHRU de Tours, Tours, France
| | | | | | - Florent Dingli
- Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, Paris, France
| | - Andrei Turtoi
- Tumor Microenvironment Laboratory, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Montpellier, France.,Institut du Cancer de Montpellier, Montpellier, France.,Université de Montpellier, Montpellier, France
| | - Audrey Le Gouellec
- CNRS, CHU Grenoble Alpes, Grenoble INP, TIMC-IMAG, University Grenoble Alpes, Grenoble, France
| | - Florence Fauvelle
- UGA/INSERM U1216, Grenoble Institute of Neurosciences, Grenoble, France.,UGA/INSERM US17, Grenoble MRI Facility IRMaGe, Grenoble, France
| | - Amélie Donchet
- Institut de Biologie Structurale (IBS), CEA, CNRS, University Grenoble Alpes, Grenoble, France
| | - Thibaut Crépin
- Institut de Biologie Structurale (IBS), CEA, CNRS, University Grenoble Alpes, Grenoble, France
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, Netherlands
| | - Christophe Paget
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Damarys Loew
- Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, PSL Research University, Paris, France
| | - Olivier Herault
- Université de Tours, Tours, France.,CNRS ERL 7001 LNOx "Leukemic niche and redox metabolism", Tours, France.,Service d'Hématologie Biologique, CHRU de Tours, Tours, France
| | - Nadia Naffakh
- Institut Pasteur, Unité Biologie des ARN et Virus Influenza, CNRS UMR3569, Paris, France
| | - Ronan Le Goffic
- Virologie et Immunologie Moléculaires, INRAe, Université Paris-Saclay, Jouy-en-Josas, France
| | - Mustapha Si-Tahar
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| |
Collapse
|
22
|
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
|
23
|
Tettamanti S, Pievani A, Biondi A, Dotti G, Serafini M. Catch me if you can: how AML and its niche escape immunotherapy. Leukemia 2022; 36:13-22. [PMID: 34302116 PMCID: PMC8727297 DOI: 10.1038/s41375-021-01350-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023]
Abstract
In spite of the remarkable progress in basic and preclinical studies of acute myeloid leukemia (AML), the five-year survival rate of AML patients remains poor, highlighting the urgent need for novel and synergistic therapies. Over the past decade, increased attention has been focused on identifying suitable immunotherapeutic strategies for AML, and in particular on targeting leukemic cells and their progenitors. However, recent studies have also underlined the important contribution of the leukemic microenvironment in facilitating tumor escape mechanisms leading to disease recurrence. Here, we describe the immunological features of the AML niche, with particular attention to the crosstalk between the AML blasts and the cellular components of the altered tumor microenvironment (TME) and the mechanisms of immune escape that hamper the therapeutic effects of the most advanced treatments. Considering the AML complexity, immunotherapy approaches may benefit from a rational combination of complementary strategies aimed at preventing escape mechanisms without increasing toxicity.
Collapse
Affiliation(s)
- Sarah Tettamanti
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | - Alice Pievani
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | - Andrea Biondi
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy.
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Marta Serafini
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| |
Collapse
|
24
|
Pimenta DB, Varela VA, Datoguia TS, Caraciolo VB, Lopes GH, Pereira WO. The Bone Marrow Microenvironment Mechanisms in Acute Myeloid Leukemia. Front Cell Dev Biol 2021; 9:764698. [PMID: 34869355 PMCID: PMC8639599 DOI: 10.3389/fcell.2021.764698] [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: 08/25/2021] [Accepted: 10/20/2021] [Indexed: 12/13/2022] Open
Abstract
Bone marrow (BM) is a highly complex tissue that provides important regulatory signals to orchestrate hematopoiesis. Resident and transient cells occupy and interact with some well characterized niches to produce molecular and cellular mechanisms that interfere with differentiation, migration, survival, and proliferation in this microenvironment. The acute myeloid leukemia (AML), the most common and severe hematological neoplasm in adults, arises and develop in the BM. The osteoblastic, vascular, and reticular niches provide surface co-receptors, soluble factors, cytokines, and chemokines that mediate important functions on hematopoietic cells and leukemic blasts. There are some evidences of how AML modify the architecture and function of these three BM niches, but it has been still unclear how essential those modifications are to maintain AML development. Basic studies and clinical trials have been suggesting that disturbing specific cells and molecules into the BM niches might be able to impair leukemia competencies. Either through niche-specific molecule inhibition alone or in combination with more traditional drugs, the bone marrow microenvironment is currently considered the potential target for new strategies to treat AML patients. This review describes the cellular and molecular constitution of the BM niches under healthy and AML conditions, presenting this anatomical compartment by a new perspective: as a prospective target for current and next generation therapies.
Collapse
Affiliation(s)
- Débora Bifano Pimenta
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Vanessa Araujo Varela
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Tarcila Santos Datoguia
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Victória Bulcão Caraciolo
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Gabriel Herculano Lopes
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Welbert Oliveira Pereira
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
25
|
Low-Dose Pesticides Alter Primary Human Bone Marrow Mesenchymal Stem/Stromal Cells through ALDH2 Inhibition. Cancers (Basel) 2021; 13:cancers13225699. [PMID: 34830855 PMCID: PMC8616329 DOI: 10.3390/cancers13225699] [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: 09/20/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 12/25/2022] Open
Abstract
(1) Background: The impact of occupational exposure to high doses of pesticides on hematologic disorders is widely studied. Yet, lifelong exposure to low doses of pesticides, and more particularly their cocktail effect, although poorly known, could also participate to the development of such hematological diseases as myelodysplastic syndrome (MDS) in elderly patients. (2) Methods: In this study, a cocktail of seven pesticides frequently present in water and food (maneb, mancozeb, iprodione, imazalil, chlorpyrifos ethyl, diazinon and dimethoate), as determined by the European Food Safety Authority, were selected. Their in vitro effects at low-doses on primary BM-MSCs from healthy volunteers were examined. (3) Results: Exposure of normal BM-MSCs to pesticides for 21 days inhibited cell proliferation and promoted DNA damage and senescence. Concomitantly, these cells presented a decrease in aldehyde dehydrogenase 2 (ALDH2: mRNA, protein and enzymatic activity) and an increase in acetaldehyde levels. Pharmacological inhibition of ALDH2 with disulfiram recapitulated the alterations induced by exposure to low doses of pesticides. Moreover, BM-MSCs capacity to support primitive hematopoiesis was significantly altered. Similar biological abnormalities were found in primary BM-MSCs derived from MDS patients. (4) Conclusions: these results suggest that ALDH2 could participate in the pathophysiology of MDS in elderly people long exposed to low doses of pesticides.
Collapse
|
26
|
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
|
27
|
Fateen M, Seif A, Salama R, Shams A, Amin D. The Relationship between the Connexin 32 and Connexin 43 Genes and the Pretreatment Stage and Short-term Follow-up of Patients with Acute Myeloid Leukemia. IRANIAN JOURNAL OF MEDICAL SCIENCES 2021; 46:347-354. [PMID: 34539009 PMCID: PMC8438339 DOI: 10.30476/ijms.2020.84511.1477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 06/13/2020] [Accepted: 08/01/2020] [Indexed: 11/19/2022]
Abstract
Background Connexins (Cxs) are gap junction proteins involved in the communication between acute myeloid leukemia (AML) and stromal cells. They consist of intercellular channels termed "connexions", which can cause uncontrolled cell proliferation if dysregulated. This study aimed to evaluate the expression levels of the Cx32 and Cx43 genes and their correlations with other prognostic markers in patients with AML. Methods This cross sectional study was performed on peripheral blood samples from 60 newly diagnosed patients with AML and 40 healthy control subjects at Kasr Alainy School of Medicine, Cairo University, from June 2016 to December 2017. The quantitative real-time polymerase chain reaction (qRT-PCR) test was used to examine the relative expression level of Cx43 and Cx32 genes in the patients and the control subjects. The Chi square test or the Fisher exact test was employed to examine the relationship between qualitative variables, while the independent t test or the Mann-Whitney test was employed for quantitative data. All the tests were two-tailed, and a P value of less than 0.05 was considered significant. Results Among the patients with AML, 65% had a high Cx32 expression level, whereas 63.3% had a low Cx43 expression level. There was a statistically significant difference in the fold change values of Cx32 and Cx43 expression between the patient group and the control group (P=0.009 vs P=0.013, respectively). There was a remarkable association between both Cxs and CD34 and HLA-DR cells. Conclusion Cx expression in samples may add to the diagnostic workup of AML. Although we found a negative correlation between Cx43 expression and the peripheral blood blast percentage, the response after the first induction of chemotherapy showed no significant relationship with Cx43 and Cx32.
Collapse
Affiliation(s)
- Mohamed Fateen
- Department of Clinical and Chemical Pathology, Cairo University, Cairo, Egypt
| | - Alia Seif
- Department of Clinical and Chemical Pathology, Cairo University, Cairo, Egypt
| | - Rasha Salama
- Department of Clinical Oncology, Cairo University, Cairo, Egypt
| | - Ahmed Shams
- Department of Clinical and Chemical Pathology, Cairo University, Cairo, Egypt
| | - Dalia Amin
- Department of Clinical and Chemical Pathology, Cairo University, Cairo, Egypt
| |
Collapse
|
28
|
Aung MMK, Mills ML, Bittencourt‐Silvestre J, Keeshan K. Insights into the molecular profiles of adult and paediatric acute myeloid leukaemia. Mol Oncol 2021; 15:2253-2272. [PMID: 33421304 PMCID: PMC8410545 DOI: 10.1002/1878-0261.12899] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/18/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022] Open
Abstract
Acute myeloid leukaemia (AML) is a clinically and molecularly heterogeneous disease characterised by uncontrolled proliferation, block in differentiation and acquired self-renewal of hematopoietic stem and myeloid progenitor cells. This results in the clonal expansion of myeloid blasts within the bone marrow and peripheral blood. The incidence of AML increases with age, and in childhood, AML accounts for 20% of all leukaemias. Whilst there are many clinical and biological similarities between paediatric and adult AML with continuum across the age range, many characteristics of AML are associated with age of disease onset. These include chromosomal aberrations, gene mutations and differentiation lineage. Following chemotherapy, AML cells that survive and result in disease relapse exist in an altered chemoresistant state. Molecular profiling currently represents a powerful avenue of experimentation to study AML cells from adults and children pre- and postchemotherapy as a means of identifying prognostic biomarkers and targetable molecular vulnerabilities that may be age-specific. This review highlights recent advances in our knowledge of the molecular profiles with a focus on transcriptomes and metabolomes, leukaemia stem cells and chemoresistant cells in adult and paediatric AML and focus on areas that hold promise for future therapies.
Collapse
Affiliation(s)
- Myint Myat Khine Aung
- Paul O’Gorman Leukaemia Research CentreInstitute of Cancer SciencesUniversity of GlasgowUK
| | - Megan L. Mills
- Paul O’Gorman Leukaemia Research CentreInstitute of Cancer SciencesUniversity of GlasgowUK
| | | | - Karen Keeshan
- Paul O’Gorman Leukaemia Research CentreInstitute of Cancer SciencesUniversity of GlasgowUK
| |
Collapse
|
29
|
Bourgeais J, Hérault O. In Vitro Analysis of Energy Metabolism in Bone-Marrow Mesenchymal Stromal Cells. Methods Mol Biol 2021; 2308:59-70. [PMID: 34057714 DOI: 10.1007/978-1-0716-1425-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Bone marrow mesenchymal stromal cells (MSCs) play an essential role in the regulation of normal and leukemic hematopoiesis. Their multipotent potential of differentiation also makes them an interesting therapeutic tool. Among factors involved in the regulation of MSCs, energy metabolism plays a key role in their proliferation and differentiation. Seahorse Bioscience introduced extracellular flux technology to the life sciences market in 2006. This methodology allows, in living cells and in real time, the concomitant determination of basal oxygen consumption, glycolysis rates, ATP production, and respiratory capacity in a single experiment. Here we describe the protocol used to study concomitantly the respiratory and glycolytic metabolism of primary MSCs from the determination of oxygen consumption (OCR) and extracellular acidification (ECAR) rates.
Collapse
Affiliation(s)
- Jérôme Bourgeais
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, Faculty of Medicine, Tours University, Tours, France.,Department of Biolocical Hematology, Tours University Hospital, Tours, France
| | - Olivier Hérault
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France. .,EA7501 GICC, Faculty of Medicine, Tours University, Tours, France. .,Department of Biolocical Hematology, Tours University Hospital, Tours, France.
| |
Collapse
|
30
|
Characterization of NADPH Oxidase Expression and Activity in Acute Myeloid Leukemia Cell Lines: A Correlation with the Differentiation Status. Antioxidants (Basel) 2021; 10:antiox10030498. [PMID: 33807114 PMCID: PMC8004739 DOI: 10.3390/antiox10030498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/14/2022] Open
Abstract
In acute myeloid leukemia (AML), a low level of reactive oxygen species (ROS) is associated with leukemic stem cell (LSC) quiescence, whereas a high level promotes blast proliferation. ROS homeostasis relies on a tightly-regulated balance between the antioxidant and oxidant systems. Among the oxidants, NADPH oxidases (NOX) generate ROS as a physiological function. Although it has been reported in AML initiation and development, the contribution of NOX to the ROS production in AML remains to be clarified. The aim of this study was to investigate the NOX expression and function in AML, and to examine the role of NOX in blast proliferation and differentiation. First, we interrogated the NOX expression in primary cells from public datasets, and investigated their association with prognostic markers. Next, we explored the NOX expression and activity in AML cell lines, and studied the impact of NOX knockdown on cell proliferation and differentiation. We found that NOX2 is ubiquitously expressed in AML blasts, and particularly in cells from the myelomonocytic (M4) and monocytic (M5) stages; however, it is less expressed in LSCs and in relapsed AML. This is consistent with an increased expression throughout normal hematopoietic differentiation, and is reflected in AML cell lines. Nevertheless, no endogenous NOX activity could be detected in the absence of PMA stimulation. Furthermore, CYBB knockdown, although hampering induced NOX2 activity, did not affect the proliferation and differentiation of THP-1 and HL-60 cells. In summary, our data suggest that NOX2 is a marker of AML blast differentiation, while AML cell lines lack any NOX2 endogenous activity.
Collapse
|
31
|
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
|
32
|
Involvement of GPx-3 in the Reciprocal Control of Redox Metabolism in the Leukemic Niche. Int J Mol Sci 2020; 21:ijms21228584. [PMID: 33202543 PMCID: PMC7696155 DOI: 10.3390/ijms21228584] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/21/2022] Open
Abstract
The bone marrow (BM) microenvironment plays a crucial role in the development and progression of leukemia (AML). Intracellular reactive oxygen species (ROS) are involved in the regulation of the biology of leukemia-initiating cells, where the antioxidant enzyme GPx-3 could be involved as a determinant of cellular self-renewal. Little is known however about the role of the microenvironment in the control of the oxidative metabolism of AML cells. In the present study, a coculture model of BM mesenchymal stromal cells (MSCs) and AML cells (KG1a cell-line and primary BM blasts) was used to explore this metabolic pathway. MSC-contact, rather than culture with MSC-conditioned medium, decreases ROS levels and inhibits the Nrf-2 pathway through overexpression of GPx3 in AML cells. The decrease of ROS levels also inactivates p38MAPK and reduces the proliferation of AML cells. Conversely, contact with AML cells modifies MSCs in that they display an increased oxidative stress and Nrf-2 activation, together with a concomitant lowered expression of GPx-3. Altogether, these experiments suggest that a reciprocal control of oxidative metabolism is initiated by direct cell–cell contact between MSCs and AML cells. GPx-3 expression appears to play a crucial role in this cross-talk and could be involved in the regulation of leukemogenesis.
Collapse
|