1
|
Hong M, Guo J, Zhao Y, Song L, Zhao S, Wang R, Shi L, Zhang Z, Wu D, He Q, Chang C. Eltrombopag restores proliferative capacity and adipose-osteogenic balance of mesenchymal stromal cells in low-risk myelodysplastic syndromes. Eur J Pharmacol 2024; 985:177086. [PMID: 39481629 DOI: 10.1016/j.ejphar.2024.177086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/13/2024] [Accepted: 10/29/2024] [Indexed: 11/02/2024]
Abstract
In low-risk myelodysplastic syndromes (MDS), the proinflammatory signaling is excessive, and the proliferation and differentiation potentials of mesenchymal stromal cells (MSCs) are strongly impaired. Eltrombopag (ELT) has been demonstrated recently effective and relatively safe in low-risk MDS with severe thrombocytopenia. However, its impact on the MDS-MSCs has not been investigated in any detail. Here, for the first time, we investigated the changes induced by ELT in MSCs' viability, proliferation, apoptosis, senescence, multilineage differentiation properties, and stem cell support capacity in low-risk MDS patients. We demonstrated that ELT may act on improving the impaired inflammatory profile and reactivating the downregulated canonical WNT signaling pathway in low-risk MDS, and also restoring the self-renewal capacity and the balance in adipose-osteogenic differentiation of MDS-MSCs.
Collapse
Affiliation(s)
- Minghua Hong
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Juan Guo
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Youshan Zhao
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Luxi Song
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Sida Zhao
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Roujia Wang
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Lei Shi
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zheng Zhang
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Dong Wu
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qi He
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chunkang Chang
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
2
|
Ceneri E, De Stefano A, Casalin I, Finelli C, Curti A, Paolini S, Parisi S, Ardizzoia F, Cristiano G, Boultwood J, McCubrey JA, Suh PG, Ramazzotti G, Fiume R, Ratti S, Manzoli L, Cocco L, Follo MY. Signaling pathways and bone marrow microenvironment in myelodysplastic neoplasms. Adv Biol Regul 2024:101071. [PMID: 39648082 DOI: 10.1016/j.jbior.2024.101071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
Key signaling pathways within the Bone Marrow Microenvironment (BMM), such as Notch, Phosphoinositide-Specific Phospholipase C (PI-PLCs), Transforming Growth Factor β (TGF-β), and Nuclear Factor Kappa B (NF-κB), play a vital role in the progression of Myelodysplastic Neoplasms (MDS). Among the various BMM cell types, Mesenchymal Stromal Cells (MSCs) are particularly central to these pathways. While these signaling routes can independently affect both MSCs and Hematopoietic Stem Cells (HSCs), they most importantly alter the dynamics of their interactions, leading to abnormal changes in survival, differentiation, and quiescence. Notch and PI-PLC signaling facilitate intercellular communication, TGF-β promotes quiescence and suppresses hematopoiesis, and NF-κB-driven inflammatory responses foster an environment detrimental to normal hematopoiesis. This review highlights the role of these pathways within the MDS microenvironment, driving the development and progression of the disease and paving the way for new possible therapeutic strategies.
Collapse
Affiliation(s)
- Eleonora Ceneri
- Department of Biomedical and Neuromotor Sciences, Cellular Signaling Laboratory, University of Bologna, Bologna, 40126, Italy.
| | - Alessia De Stefano
- Department of Biomedical and Neuromotor Sciences, Cellular Signaling Laboratory, University of Bologna, Bologna, 40126, Italy
| | - Irene Casalin
- Department of Biomedical and Neuromotor Sciences, Cellular Signaling Laboratory, University of Bologna, Bologna, 40126, Italy
| | - Carlo Finelli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seragnoli", Bologna, Italy
| | - Antonio Curti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seragnoli", Bologna, Italy
| | - Stefania Paolini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seragnoli", Bologna, Italy
| | - Sarah Parisi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seragnoli", Bologna, Italy
| | - Federica Ardizzoia
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seragnoli", Bologna, Italy
| | - Gianluca Cristiano
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seragnoli", Bologna, Italy
| | - Jaqueline Boultwood
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Pann-Ghill Suh
- Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Giulia Ramazzotti
- Department of Biomedical and Neuromotor Sciences, Cellular Signaling Laboratory, University of Bologna, Bologna, 40126, Italy
| | - Roberta Fiume
- Department of Biomedical and Neuromotor Sciences, Cellular Signaling Laboratory, University of Bologna, Bologna, 40126, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Cellular Signaling Laboratory, University of Bologna, Bologna, 40126, Italy
| | - Lucia Manzoli
- Department of Biomedical and Neuromotor Sciences, Cellular Signaling Laboratory, University of Bologna, Bologna, 40126, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Cellular Signaling Laboratory, University of Bologna, Bologna, 40126, Italy
| | - Matilde Y Follo
- Department of Biomedical and Neuromotor Sciences, Cellular Signaling Laboratory, University of Bologna, Bologna, 40126, Italy
| |
Collapse
|
3
|
Zhang Y, Ren J, Liao Z, Li X, Zhang C, Huang B, Cao Y, Chen J. Downregulating LKB1 in bone marrow mesenchymal stem cells could inhibit CD4 + T cell proliferation via the PD-1/PD-L1 signaling pathway. Immunobiology 2024; 229:152856. [PMID: 39369651 DOI: 10.1016/j.imbio.2024.152856] [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: 05/09/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Our previous research has shown that LKB1 in amniotic mesenchymal stem cells (MSCs) serves as a vital regulator of regulatory T cell differentiation and T cell proliferation, which may have a similar role in bone marrow MSCs (BMMSCs). Therefore, we investigated the role of LKB1 in BMMSCs for regulating CD4+ T cell proliferation in the bone micro-environment of AML. METHODS RT-PCR was used to assessed LKB1 expression in BMMSCs derived from AML patients and healthy controls. Subsequently, LKB1 was knocked down in the BMMSCs line HS-5 (HS-5-LKB1KD). Co-cultures in vitro were established to analyze the effect of HS-5-LKB1KD on CD4+ T cell. Flow cytometry was employed to measure PD-L1 and CD4+ T cell proliferation levels. Western blot was utilized to detect related proteins. RESULTS The expression of LKB1 in BMMSCs derived from AML patients was decreased. Knockdown of LKB1 in HS-5 resulted in upregulation of PD-L1 expression. Co-culture of peripheral blood CD4+ T cell with HS-5-LKB1KD exhibited reduced CD4+ T cell proliferation compared to co-culture with HS-5-LKB1con. Furthermore, blocking PD-L1 in the co-culture conditions could restore the reduced CD4+ T cell proliferation. Additionally, it was found that upregulation of the Wnt signaling pathway-related proteins following LKB1 knockdown in HS-5, indicating that downregulating LKB1 could promote PD-L1 expression through activation of the Wnt signaling pathway. CONCLUSIONS The decreased expression of LKB1 in BMMSCs may activate the Wnt signaling pathway, leading to increased PD-L1 expression. This inhibited CD4+ T cell proliferation, which might lead to impaired anti-tumor immunity in AML patients and promote AML progression.
Collapse
Affiliation(s)
- Yaqin Zhang
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Jingyi Ren
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Zhongxian Liao
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Xiaoyu Li
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Chunying Zhang
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Bihan Huang
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China
| | - Yingping Cao
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China.
| | - Jiadi Chen
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350004, PR China.
| |
Collapse
|
4
|
Landspersky T, Stein M, Saçma M, Geuder J, Braitsch K, Rivière J, Hettler F, Romero Marquez S, Vilne B, Hameister E, Richter D, Schönhals E, Tuckermann J, Verbeek M, Herhaus P, Hecker JS, Bassermann F, Götze KS, Enard W, Geiger H, Oostendorp RAJ, Schreck C. Targeting CDC42 reduces skeletal degeneration after hematopoietic stem cell transplantation. Blood Adv 2024; 8:5400-5414. [PMID: 39159429 PMCID: PMC11526086 DOI: 10.1182/bloodadvances.2024012879] [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: 02/07/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 08/21/2024] Open
Abstract
ABSTRACT Osteopenia and osteoporosis are common long-term complications of the cytotoxic conditioning regimen for hematopoietic stem cell transplantation (HSCT). We examined mesenchymal stem and progenitor cells (MSPCs), which include skeletal progenitors, from mice undergoing HSCT. Such MSPCs showed reduced fibroblastic colony-forming units frequency, increased DNA damage, and enhanced occurrence of cellular senescence, whereas there was a reduced bone volume in animals that underwent HSCT. This reduced MSPC function correlated with elevated activation of the small Rho guanosine triphosphate hydrolase CDC42, disorganized F-actin distribution, mitochondrial abnormalities, and impaired mitophagy in MSPCs. Changes and defects similar to those in mice were also observed in MSPCs from humans undergoing HSCT. A pharmacological treatment that attenuated the elevated activation of CDC42 restored F-actin fiber alignment, mitochondrial function, and mitophagy in MSPCs in vitro. Finally, targeting CDC42 activity in vivo in animals undergoing transplants improved MSPC quality to increase both bone volume and trabecular bone thickness. Our study shows that attenuation of CDC42 activity is sufficient to attenuate reduced function of MSPCs in a BM transplant setting.
Collapse
Affiliation(s)
- Theresa Landspersky
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Merle Stein
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Mehmet Saçma
- Institute of Molecular Medicine, Stem Cells, and Aging, Ulm University, Ulm, Germany
| | - Johanna Geuder
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians University, Munich, Germany
| | - Krischan Braitsch
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Jennifer Rivière
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Franziska Hettler
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Sandra Romero Marquez
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Baiba Vilne
- Bioinformatics Laboratory, Rīga Stradiņš University, Riga, Lettland
| | - Erik Hameister
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Daniel Richter
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Emely Schönhals
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Mareike Verbeek
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Peter Herhaus
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Judith S. Hecker
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Florian Bassermann
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Katharina S. Götze
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians University, Munich, Germany
| | - Hartmut Geiger
- Institute of Molecular Medicine, Stem Cells, and Aging, Ulm University, Ulm, Germany
| | - Robert A. J. Oostendorp
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Christina Schreck
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| |
Collapse
|
5
|
Xue C, Chen L, Wang N, Chen H, Xu W, Xi Z, Sun Q, Kang R, Xie L, Liu X. Stimuli-responsive hydrogels for bone tissue engineering. BIOMATERIALS TRANSLATIONAL 2024; 5:257-273. [PMID: 39734705 PMCID: PMC11681187 DOI: 10.12336/biomatertransl.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/22/2024] [Accepted: 08/30/2024] [Indexed: 12/31/2024]
Abstract
The treatment of bone defects remains a great clinical challenge. With the development of science and technology, bone tissue engineering technology has emerged, which can mimic the structure and function of natural bone tissues and create solutions for repairing or replacing human bone tissues based on biocompatible materials, cells and bioactive factors. Hydrogels are favoured by researchers due to their high water content, degradability and good biocompatibility. This paper describes the hydrogel sources, roles and applications. According to the different types of stimuli, hydrogels are classified into three categories: physical, chemical and biochemical responses, and the applications of different stimuli-responsive hydrogels in bone tissue engineering are summarised. Stimuli-responsive hydrogels can form a semi-solid with good adhesion based on different physiological environments, which can carry a variety of bone-enhancing bioactive factors, drugs and cells, and have a long retention time in the local area, which is conducive to a long period of controlled release; they can also form a scaffold for constructing tissue repair, which can jointly promote the repair of bone injury sites. However, it also has many defects, such as poor biocompatibility, immunogenicity and mechanical stability. Further studies are still needed in the future to facilitate its clinical translation.
Collapse
Affiliation(s)
- Congyang Xue
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Liping Chen
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Nan Wang
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Heng Chen
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Wenqiang Xu
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Zhipeng Xi
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Qing Sun
- Laboratory of Gene Therapy, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Ran Kang
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Lin Xie
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Xin Liu
- Department of Orthopaedics, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| |
Collapse
|
6
|
Wang W, Brown TJ, Barth BM. Influences on the Hematopoietic Stem Cell Niche. SCIBASE HEMATOLOGY & BLOOD DISORDERS 2024; 1:1002. [PMID: 39429505 PMCID: PMC11486556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Hematopoietic stem cells (HSCs) are supported by the bone marrow microenvironment to maintain normal production of blood cells. The niche may be considered an "ecosystem" that support the function of HSCs and other supportive cells. Alterations in the bone marrow niche are commonly observed in hematologic malignancies. Here, we review recent insights into the location and the molecular and cellular components of the bone marrow niche. Moreover, we discuss how the niche interacts with HSCs to drive the pathogenesis of hematopoietic malignancies. Overall, a better understanding of the influences on the HSC niche may drive therapeutic development targeting defective and aberrant hematopoiesis.
Collapse
Affiliation(s)
- Weiyuan Wang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta GA 30322 USA
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham NH 03824 USA
| | - Timothy J. Brown
- Division of Hematology and Oncology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas TX 75390 USA
| | - Brian M. Barth
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham NH 03824 USA
- Department of Natural Sciences, University of Alaska Southeast, Juneau AK 99801 USA
| |
Collapse
|
7
|
Trivanović D, Vujačić M, Labella R, Djordjević IO, Ćazić M, Chernak B, Jauković A. Molecular Deconvolution of Bone Marrow Adipose Tissue Interactions with Malignant Hematopoiesis: Potential for New Therapy Development. Curr Osteoporos Rep 2024; 22:367-377. [PMID: 38922359 DOI: 10.1007/s11914-024-00879-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
PURPOSE OF REVIEW Along with a strong impact on skeletal integrity, bone marrow adipose tissue (BMAT) is an important modulator of the adult hematopoietic system. This review will summarize the current knowledge on the causal relationship between bone marrow (BM) adipogenesis and the development and progression of hematologic malignancies. RECENT FINDINGS BM adipocytes (BMAds) support a number of processes promoting oncogenesis, including the evolution of clonal hematopoiesis, malignant cell survival, proliferation, angiogenesis, and chemoresistance. In addition, leukemic cells manipulate surrounding BMAds by promoting lipolysis and release of free fatty acids, which are then utilized by leukemic cells via β-oxidation. Therefore, limiting BM adipogenesis, blocking BMAd-derived adipokines, or lipid metabolism obstruction have been considered as potential treatment options for hematological malignancies. Leukemic stem cells rely heavily on BMAds within the structural BM microenvironment for necessary signals which foster disease progression. Further development of 3D constructs resembling BMAT at different skeletal regions are critical to better understand these relationships in geometric space and may provide essential insight into the development of hematologic malignancies within the BM niche. In turn, these mechanisms provide promising potential as novel approaches to targeting the microenvironment with new therapeutic strategies.
Collapse
Affiliation(s)
- Drenka Trivanović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000, Belgrade, Serbia.
| | - Marko Vujačić
- Institute for Orthopedy Banjica, 11000, Belgrade, Serbia
- School of Medicine, University of Belgrade, 11000, Belgrade, Serbia
| | - Rossella Labella
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
- Edward P. Evans Center for Myelodysplastic Syndromes, Columbia University Medical Center, New York, NY, USA
| | - Ivana Okić Djordjević
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000, Belgrade, Serbia
| | - Marija Ćazić
- Department of Hematology and Oncology, University Children's Hospital Tiršova, 11000, Belgrade, Serbia
| | - Brian Chernak
- Division of Hematology/Oncology, Columbia University, New York, NY, USA
| | - Aleksandra Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000, Belgrade, Serbia
| |
Collapse
|
8
|
Vukotić M, Kapor S, Simon F, Cokic V, Santibanez JF. Mesenchymal stromal cells in myeloid malignancies: Immunotherapeutic opportunities. Heliyon 2024; 10:e25081. [PMID: 38314300 PMCID: PMC10837636 DOI: 10.1016/j.heliyon.2024.e25081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/06/2024] Open
Abstract
Myeloid malignancies are clonal disorders of the progenitor cells or hematopoietic stem cells, including acute myeloid leukemia, myelodysplastic syndromes, myeloproliferative malignancies, and chronic myelomonocytic leukemia. Myeloid neoplastic cells affect the proliferation and differentiation of other hematopoietic lineages in the bone marrow and peripheral blood, leading to severe and life-threatening complications. Mesenchymal stromal cells (MSCs) residing in the bone marrow exert immunosuppressive functions by suppressing innate and adaptive immune systems, thus creating a supportive and tolerant microenvironment for myeloid malignancy progression. This review summarizes the significant features of MSCs in myeloid malignancies, including their role in regulating cell growth, cell death, and antineoplastic resistance, in addition to their immunosuppressive contributions. Understanding the implications of MSCs in myeloid malignancies could pave the path for potential use in immunotherapy.
Collapse
Affiliation(s)
- Milica Vukotić
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Suncica Kapor
- Department of Hematology, Clinical Hospital Center “Dr. Dragisa Misovic-Dedinje,” University of Belgrade, Serbia
| | - Felipe Simon
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Millennium Nucleus of Ion Channel-Associated Diseases, Universidad de Chile, Santiago, Chile
| | - Vladan Cokic
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Juan F. Santibanez
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| |
Collapse
|
9
|
Wang Q, Liu J, Zhong Y, Li D, Zhong Y, Ying H, Zhang T. A Fanca knockout mouse model reveals novel Fancd2 function. Biochem Biophys Res Commun 2024; 696:149454. [PMID: 38217981 DOI: 10.1016/j.bbrc.2023.149454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/10/2023] [Accepted: 12/27/2023] [Indexed: 01/15/2024]
Abstract
Fanconi anemia (FA) is a genetically and clinically heterogenous inherited disorder. Clinically, Fanca subtype patients exhibited milder phenotypes compared to Fancd2 subtypes. Increasing evidence suggests that Fancd2 perform independent functions, but the detailed mechanisms are not well characterized. In this study, we developed a Fanca KO mice model in C57BL/6 background with ATG region deletion, then performed a detailed FA phenotypes characterization and analysis with Fanca KO mice and Fancd2 KO mice in the same congenic background. We found that both the Fanca KO and Fancd2 KO cause severe FA phenotypes in mice. However, Fanca KO mice exhibited milder FA phenotypes comparing to Fancd2 KO mice. Fanca KO mice showed higher embryonic and postnatal survival rate, less congenital eye defects in early development. At adult stage, Fanca KO mice showed increased HSC number and reconstitution function. Furthermore, we did RNA-seq study and identified differential expression of Dlk1 and Dlk1 pathway genes in Fanca KO and Fancd2 KO embryonic cells and adult HSCs. Finally, we revealed that Fancd2 was expressed and physically interact with Dlk1 in Fanca KO cells. Collectively, our findings suggested that Fancd2 has distinct functions in the absence of Fanca.
Collapse
Affiliation(s)
- Qian Wang
- Experimental Animal Research Center, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jia Liu
- Experimental Animal Research Center, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yixinhe Zhong
- Experimental Animal Research Center, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Dongbo Li
- Experimental Animal Research Center, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yusen Zhong
- Experimental Animal Research Center, Hangzhou Medical College, Hangzhou, Zhejiang, China; Zhejiang Provincial Laboratory of Experimental Animals & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Huazhong Ying
- Experimental Animal Research Center, Hangzhou Medical College, Hangzhou, Zhejiang, China; Zhejiang Provincial Laboratory of Experimental Animals & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Tingting Zhang
- Experimental Animal Research Center, Hangzhou Medical College, Hangzhou, Zhejiang, China; Zhejiang Provincial Laboratory of Experimental Animals & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
10
|
Singh AK, Prasad P, Cancelas JA. Mesenchymal stromal cells, metabolism, and mitochondrial transfer in bone marrow normal and malignant hematopoiesis. Front Cell Dev Biol 2023; 11:1325291. [PMID: 38169927 PMCID: PMC10759248 DOI: 10.3389/fcell.2023.1325291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
Hematopoietic stem cell (HSC) transplantation-based treatments are in different phases of clinical development, ranging from current therapies to a promise in the repair and regeneration of diseased tissues and organs. Mesenchymal stromal/stem cells (MSCs), which are fibroblast-like heterogeneous progenitors with multilineage differentiation (osteogenic, chondrogenic, and adipogenic) and self-renewal potential, and exist in the bone marrow (BM), adipose, and synovium, among other tissues, represent one of the most widely used sources of stem cells in regenerative medicine. MSCs derived from bone marrow (BM-MSCs) exhibit a variety of traits, including the potential to drive HSC fate and anti-inflammatory and immunosuppressive capabilities via paracrine activities and interactions with the innate and adaptive immune systems. The role of BM-MSC-derived adipocytes is more controversial and may act as positive or negative regulators of benign or malignant hematopoiesis based on their anatomical location and functional crosstalk with surrounding cells in the BM microenvironment. This review highlights the most recent clinical and pre-clinical findings on how BM-MSCs interact with the surrounding HSCs, progenitors, and immune cells, and address some recent insights on the mechanisms that mediate MSCs and adipocyte metabolic control through a metabolic crosstalk between BM microenvironment cells and intercellular mitochondrial transfer in normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Abhishek K. Singh
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Parash Prasad
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Jose A. Cancelas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
11
|
Wan Z, zhang Y, Lv J, Yuan Y, Guo W, Leng Y. Exosomes derived from bone marrow mesenchymal stem cells regulate pyroptosis via the miR-143-3p/myeloid differentiation factor 88 axis to ameliorate intestinal ischemia-reperfusion injury. Bioengineered 2023; 14:2253414. [PMID: 37674357 PMCID: PMC10486297 DOI: 10.1080/21655979.2023.2253414] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/02/2022] [Accepted: 08/20/2022] [Indexed: 09/08/2023] Open
Abstract
Intestinal ischemia-reperfusion (I/R) injury is a condition in which tissue injury is aggravated after ischemia due to recovery of blood supply. Bone marrow mesenchymal stem cell-derived exosome (BMSC-exo) showed a protective effect on I/R injury. This study aimed to investigate the possible mechanisms by which BMSC-exos ameliorate intestinal I/R injury. We isolated mouse BMSC-exos by super-centrifugation and found that they effectively increased cell viability in a cell model, alleviated intestinal barrier injury in a mouse model, and downregulated the expression of inflammatory cytokines and pyroptosis-related proteins, suggesting that BMSC-exos may alleviate intestinal I/R injury in vitro and in vivo by regulating pyroptosis. We identified miR-143-3p as a differentially expressed miRNA by microarray sequencing. Bioinformatic analysis predicted a binding site between miR-143-3p and myeloid differentiation factor 88 (MyD88); a dual-luciferase reporter assay confirmed that miR-143-3p could directly regulate the expression of MyD88. Our findings suggest that miR-143-3p regulates pyroptosis by regulating NOD-like receptor thermal protein domain associated protein 3 (NLRP3) through the toll-like receptor (TLR)-4/MyD88/nuclear factor kappa-B (NF-кB) pathway. This study describes a potential strategy for the treatment of intestinal I/R injury using BMSC-exos that act by regulating pyroptosis through the miR-143-3p mediated TLR4/MyD88/NF-кB pathway.
Collapse
Affiliation(s)
- Zhanhai Wan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Yan zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Jipeng Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Yuan Yuan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Wenwen Guo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Yufang Leng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| |
Collapse
|
12
|
Li H, Wang Y, Yang F, Feng S, Chang K, Yu X, Guan F, Li X. Clonal MDS/AML cells with enhanced TWIST1 expression reprogram the differentiation of bone marrow MSCs. Redox Biol 2023; 67:102900. [PMID: 37748319 PMCID: PMC10520935 DOI: 10.1016/j.redox.2023.102900] [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: 07/23/2023] [Revised: 09/11/2023] [Accepted: 09/20/2023] [Indexed: 09/27/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMMSCs) derived from myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) patients often show a shift in the balance between osteoblastogenesis and adipogenesis. It was suggested that BMMSCs can potentially undergo reprogramming or educational processes. However, the results of reprogrammed differentiation have been inconclusive. In this study, clinical samples, co-culture models and mouse models were employed to explore the association of MDS/AML clonal cells and BMMSCs differentiation. We found that clonal MDS/AML cells promoted adipogenic differentiation and inhibited osteogenic differentiation of BMMSCs, which in turn promoted MDS expansion. Mass spectrometry and cytokine array were used to identify the molecules to drive the BMMSCs differentiation in MDS/AML. Mechanistically, highly expressed transcription factor TWIST1 in clonal MDS/AML cells induces MDS/AML cells to secrete more IFN-γ, which can induce oxidative stress through STAT1-dependent manner, ultimately causing enhanced adipogenic differentiation and inhibited osteogenic differentiation in BMMSCs. Overall, our findings suggest that targeting the driving oncogenes in malignant clonal cells, such as TWIST1, may offer new therapeutic strategies by remodeling the surrounding bone marrow microenvironment in the treatment of MDS/AML and other hematopoietic malignancies.
Collapse
Affiliation(s)
- Hongjiao Li
- Key Laboratory of Resource Biology and Biotechnology of Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Yi Wang
- Department of Hematology, Provincial People's Hospital, Xi'an, China
| | - Fenfang Yang
- Key Laboratory of Resource Biology and Biotechnology of Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Shuang Feng
- Key Laboratory of Resource Biology and Biotechnology of Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Kaijing Chang
- Key Laboratory of Resource Biology and Biotechnology of Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Xinwen Yu
- Key Laboratory of Resource Biology and Biotechnology of Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Feng Guan
- Key Laboratory of Resource Biology and Biotechnology of Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Xiang Li
- Institute of Hematology, School of Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
13
|
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
|
14
|
Pontikoglou CG, Matheakakis A, Papadaki HA. The mesenchymal compartment in myelodysplastic syndrome: Its role in the pathogenesis of the disorder and its therapeutic targeting. Front Oncol 2023; 13:1102495. [PMID: 36761941 PMCID: PMC9907728 DOI: 10.3389/fonc.2023.1102495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/06/2023] [Indexed: 01/26/2023] Open
Abstract
Myelodysplastic syndromes include a broad spectrum of malignant myeloid disorders that are characterized by dysplastic ineffective hematopoiesis, reduced peripheral blood cells counts and a high risk of progression to acute myeloid leukemia. The disease arises primarily because of accumulating chromosomal, genetic and epigenetic changes as well as immune-mediated alterations of the hematopoietic stem cells (HSCs). However, mounting evidence suggests that aberrations within the bone marrow microenvironment critically contribute to myelodysplastic syndrome (MDS) initiation and evolution by providing permissive cues that enable the abnormal HSCs to grow and eventually establish and propagate the disease. Mesenchymal stromal cells (MSCs) are crucial elements of the bone marrow microenvironment that play a key role in the regulation of HSCs by providing appropriate signals via soluble factors and cell contact interactions. Given their hematopoiesis supporting capacity, it has been reasonable to investigate MSCs' potential involvement in MDS. This review discusses this issue by summarizing existing findings obtained by in vitro studies and murine disease models of MDS. Furthermore, the theoretical background of targeting the BM-MSCs in MDS is outlined and available therapeutic modalities are described.
Collapse
Affiliation(s)
- Charalampos G. Pontikoglou
- Department of Hematology, School of Medicine, University of Crete, Heraklion, Greece,Haemopoiesis Research Laboratory, School of Medicine, University of Crete, Heraklion, Greece,*Correspondence: Charalampos G. Pontikoglou,
| | - Angelos Matheakakis
- Department of Hematology, School of Medicine, University of Crete, Heraklion, Greece,Haemopoiesis Research Laboratory, School of Medicine, University of Crete, Heraklion, Greece
| | - Helen A. Papadaki
- Department of Hematology, School of Medicine, University of Crete, Heraklion, Greece,Haemopoiesis Research Laboratory, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
15
|
Jiang C, Li J, Guo D, Luo Z. Bone Marrow Mesenchymal Stem Cell-Derived miR-29b Promotes the Progression of Acute Myeloid Leukemia. J BIOMATER TISS ENG 2023. [DOI: 10.1166/jbt.2023.3213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
This study investigates the effect of low miR-29b expression derived from bone marrow mesenchymal stem cells (BMSC) on AML. miR-29b expression in acute leukemia drug resistant cell line as K562/ADM was detected with RT-PCR. Cell proliferation was tested with MTT assay and apoptosis
was analyzed by flow cytometry. The correlation between miR-29b and PDGFRα level was analyzed. miR-29b expression was reduced after si-miR-29b transfection. PDGFRα expression was increased by the low miR-29b expression in AML cells so as to prompt the progression
of AML. Cell proliferation in K562/ADM was increased after miR-29b expression was reduced and quantity of apoptosis was decreased. There was a correlation of miR-29b and PDGFRα in the staging of AML (P <0.05). In conclusion, AML could be aggravated by the low miR-29b
expression possibly through regulating PDGFRα, resulting in increased drug tolerance.
Collapse
Affiliation(s)
- Chunyan Jiang
- Department of Pediatrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang, 163001, China
| | - Jinlan Li
- Department of Pediatrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang, 163001, China
| | - Dandan Guo
- Department of Pediatrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang, 163001, China
| | - Zhihong Luo
- Department of Pediatrics, Daqing Oilfield General Hospital, Daqing, Heilongjiang, 163001, China
| |
Collapse
|
16
|
Lang JB, Buck MC, Rivière J, Stambouli O, Sachenbacher K, Choudhary P, Dietz H, Giebel B, Bassermann F, Oostendorp RAJ, Götze KS, Hecker JS. Comparative analysis of extracellular vesicle isolation methods from human AML bone marrow cells and AML cell lines. Front Oncol 2022; 12:949261. [PMID: 36263223 PMCID: PMC9574064 DOI: 10.3389/fonc.2022.949261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular crosstalk between hematopoietic stem/progenitor cells and the bone marrow (BM) niche is vital for the development and maintenance of myeloid malignancies. These compartments can communicate via bidirectional transfer of extracellular vesicles (EVs). EV trafficking in acute myeloid leukemia (AML) plays a crucial role in shaping the BM microenvironment into a leukemia-permissive niche. Although several EV isolation methods have been developed, it remains a major challenge to define the most accurate and reliable procedure. Here, we tested the efficacy and functional assay compatibility of four different EV isolation methods in leukemia-derived EVs: (1) membrane affinity-based: exoEasy Kit alone and (2) in combination with Amicon filtration; (3) precipitation: ExoQuick-TC; and (4) ultracentrifugation (UC). Western blot analysis of EV fractions showed the highest enrichment of EV marker expression (e.g., CD63, HSP70, and TSG101) by precipitation with removal of overabundant soluble proteins [e.g., bovine serum albumin (BSA)], which were not discarded using UC. Besides the presence of damaged EVs after UC, intact EVs were successfully isolated with all methods as evidenced by highly maintained spherical- and cup-shaped vesicles in transmission electron microscopy. Nanoparticle tracking analysis of EV particle size and concentration revealed significant differences in EV isolation efficacy, with exoEasy Kit providing the highest EV yield recovery. Of note, functional assays with exoEasy Kit-isolated EVs showed significant toxicity towards treated target cells [e.g., mesenchymal stromal cells (MSCs)], which was abrogated when combining exoEasy Kit with Amicon filtration. Additionally, MSC treated with green fluorescent protein (GFP)-tagged exoEasy Kit-isolated EVs did not show any EV uptake, while EV isolation by precipitation demonstrated efficient EV internalization. Taken together, the choice of EV isolation procedure significantly impacts the yield and potential functionality of leukemia-derived EVs. The cheapest method (UC) resulted in contaminated and destructed EV fractions, while the isolation method with the highest EV yield (exoEasy Kit) appeared to be incompatible with functional assays. We identified two methods (precipitation-based ExoQuick-TC and membrane affinity-based exoEasy Kit combined with Amicon filtration) yielding pure and intact EVs, also suitable for application in functional assays. This study highlights the importance of selecting the right EV isolation method depending on the desired experimental design.
Collapse
Affiliation(s)
- Jonas B. Lang
- Department of Medicine III, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Michèle C. Buck
- Department of Medicine III, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Jennifer Rivière
- Department of Medicine III, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Oumaima Stambouli
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Ken Sachenbacher
- Department of Physics, Technical University of Munich (TUM), Munich, Germany
- Munich Institute of Biomedical Engineering, Technical University of Munich (TUM), Munich, Germany
| | - Purva Choudhary
- Department of Medicine III, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Hendrik Dietz
- Department of Physics, Technical University of Munich (TUM), Munich, Germany
- Munich Institute of Biomedical Engineering, Technical University of Munich (TUM), Munich, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Florian Bassermann
- Department of Medicine III, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich (TUM), Munich, Germany
| | - Robert A. J. Oostendorp
- Department of Medicine III, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Katharina S. Götze
- Department of Medicine III, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Judith S. Hecker
- Department of Medicine III, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
- *Correspondence: Judith S. Hecker,
| |
Collapse
|
17
|
Chen Y, Li J, Xu L, Găman MA, Zou Z. The genesis and evolution of acute myeloid leukemia stem cells in the microenvironment: From biology to therapeutic targeting. Cell Death Discov 2022; 8:397. [PMID: 36163119 PMCID: PMC9513079 DOI: 10.1038/s41420-022-01193-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/12/2022] [Accepted: 09/16/2022] [Indexed: 11/09/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by cytogenetic and genomic alterations. Up to now, combination chemotherapy remains the standard treatment for leukemia. However, many individuals diagnosed with AML develop chemotherapeutic resistance and relapse. Recently, it has been pointed out that leukemic stem cells (LSCs) are the fundamental cause of drug resistance and AML relapse. LSCs only account for a small subpopulation of all leukemic cells, but possess stem cell properties, including a self-renewal capacity and a multi-directional differentiation potential. LSCs reside in a mostly quiescent state and are insensitive to chemotherapeutic agents. When LSCs reside in a bone marrow microenvironment (BMM) favorable to their survival, they engage into a steady, continuous clonal evolution to better adapt to the action of chemotherapy. Most chemotherapeutic drugs can only eliminate LSC-derived clones, reducing the number of leukemic cells in the BM to a normal range in order to achieve complete remission (CR). LSCs hidden in the BM niche can hardly be targeted or eradicated, leading to drug resistance and AML relapse. Understanding the relationship between LSCs, the BMM, and the generation and evolution laws of LSCs can facilitate the development of effective therapeutic targets and increase the efficiency of LSCs elimination in AML.
Collapse
Affiliation(s)
- Yongfeng Chen
- Department of Basic Medical Sciences, Medical College of Taizhou University, Taizhou, Zhejiang, 318000, China.
| | - Jing Li
- Department of Histology and Embryology, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Linglong Xu
- Department of Hematology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, 318000, China
| | - Mihnea-Alexandru Găman
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 050474, Bucharest, Romania.
- Department of Hematology, Centre of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, Bucharest, Romania.
| | - Zhenyou Zou
- Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou, Guangxi, 545005, China.
| |
Collapse
|
18
|
Bains AK, Behrens Wu L, Rivière J, Rother S, Magno V, Friedrichs J, Werner C, Bornhäuser M, Götze KS, Cross M, Platzbecker U, Wobus M. Bone marrow mesenchymal stromal cell-derived extracellular matrix displays altered glycosaminoglycan structure and impaired functionality in Myelodysplastic Syndromes. Front Oncol 2022; 12:961473. [PMID: 36158640 PMCID: PMC9492883 DOI: 10.3389/fonc.2022.961473] [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: 06/04/2022] [Accepted: 08/22/2022] [Indexed: 11/24/2022] Open
Abstract
Myelodysplastic syndromes (MDS) comprise a heterogeneous group of hematologic malignancies characterized by clonal hematopoiesis, one or more cytopenias such as anemia, neutropenia, or thrombocytopenia, abnormal cellular maturation, and a high risk of progression to acute myeloid leukemia. The bone marrow microenvironment (BMME) in general and mesenchymal stromal cells (MSCs) in particular contribute to both the initiation and progression of MDS. However, little is known about the role of MSC-derived extracellular matrix (ECM) in this context. Therefore, we performed a comparative analysis of in vitro deposited MSC-derived ECM of different MDS subtypes and healthy controls. Atomic force microscopy analyses demonstrated that MDS ECM was significantly thicker and more compliant than those from healthy MSCs. Scanning electron microscopy showed a dense meshwork of fibrillar bundles connected by numerous smaller structures that span the distance between fibers in MDS ECM. Glycosaminoglycan (GAG) structures were detectable at high abundance in MDS ECM as white, sponge-like arrays on top of the fibrillar network. Quantification by Blyscan assay confirmed these observations, with higher concentrations of sulfated GAGs in MDS ECM. Fluorescent lectin staining with wheat germ agglutinin and peanut agglutinin demonstrated increased deposition of N-acetyl-glucosamine GAGs (hyaluronan (HA) and heparan sulfate) in low risk (LR) MDS ECM. Differential expression of N-acetyl-galactosamine GAGs (chondroitin sulfate, dermatan sulfate) was observed between LR- and high risk (HR)-MDS. Moreover, increased amounts of HA in the matrix of MSCs from LR-MDS patients were found to correlate with enhanced HA synthase 1 mRNA expression in these cells. Stimulation of mononuclear cells from healthy donors with low molecular weight HA resulted in an increased expression of various pro-inflammatory cytokines suggesting a contribution of the ECM to the inflammatory BMME typical of LR-MDS. CD34+ hematopoietic stem and progenitor cells (HSPCs) displayed an impaired differentiation potential after cultivation on MDS ECM and modified morphology accompanied by decreased integrin expression which mediate cell-matrix interaction. In summary, we provide evidence for structural alterations of the MSC-derived ECM in both LR- and HR-MDS. GAGs may play an important role in this remodeling processes during the malignant transformation which leads to the observed disturbance in the support of normal hematopoiesis.
Collapse
Affiliation(s)
- Amanpreet Kaur Bains
- Medical Department I, Haematology and Cell Therapy, University of Leipzig Medical Center, Leipzig, Germany
| | - Lena Behrens Wu
- Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Jennifer Rivière
- Department of Medicine III, Hematology/Oncology, School of Medicine, Klinikum rechts der Isar, München, Technical University of Munich, Munich, Germany
| | - Sandra Rother
- Center for Molecular Signaling Präklinisches Zentrum für Molekulare Signalverarbeitung (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Valentina Magno
- Max Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research Dresden, Technische Universität (TU) Dresden, Dresden, Germany
| | - Jens Friedrichs
- Max Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research Dresden, Technische Universität (TU) Dresden, Dresden, Germany
| | - Carsten Werner
- Max Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research Dresden, Technische Universität (TU) Dresden, Dresden, Germany
| | - Martin Bornhäuser
- Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Katharina S. Götze
- Department of Medicine III, Hematology/Oncology, School of Medicine, Klinikum rechts der Isar, München, Technical University of Munich, Munich, Germany
| | - Michael Cross
- Medical Department I, Haematology and Cell Therapy, University of Leipzig Medical Center, Leipzig, Germany
| | - Uwe Platzbecker
- Medical Department I, Haematology and Cell Therapy, University of Leipzig Medical Center, Leipzig, Germany
| | - Manja Wobus
- Department of Medicine I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
- *Correspondence: Manja Wobus,
| |
Collapse
|
19
|
Granata V, Crisafulli L, Nastasi C, Ficara F, Sobacchi C. Bone Marrow Niches and Tumour Cells: Lights and Shadows of a Mutual Relationship. Front Immunol 2022; 13:884024. [PMID: 35603212 PMCID: PMC9121377 DOI: 10.3389/fimmu.2022.884024] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/12/2022] [Indexed: 12/23/2022] Open
Abstract
The bone marrow (BM) niche is the spatial structure within the intra-trabecular spaces of spongious bones and of the cavity of long bones where adult haematopoietic stem cells (HSCs) maintain their undifferentiated and cellular self-renewal state through the intervention of vascular and nervous networks, metabolic pathways, transcriptional and epigenetic regulators, and humoral signals. Within the niche, HSCs interact with various cell types such as osteoblasts, endothelial cells, macrophages, and mesenchymal stromal cells (MSCs), which maintain HSCs in a quiescent state or sustain their proliferation, differentiation, and trafficking, depending on body needs. In physiological conditions, the BM niche permits the daily production of all the blood and immune cells and their admittance/ingress/progression into the bloodstream. However, disruption of this delicate microenvironment promotes the initiation and progression of malignancies such as those included in the spectrum of myeloid neoplasms, also favouring resistance to pharmacological therapies. Alterations in the MSC population and in the crosstalk with HSCs owing to tumour-derived factors contribute to the formation of a malignant niche. On the other hand, cells of the BM microenvironment cooperate in creating a unique milieu favouring metastasization of distant tumours into the bone. In this framework, the pro-tumorigenic role of MSCs is well-documented, and few evidence suggest also an anti-tumorigenic effect. Here we will review recent advances regarding the BM niche composition and functionality in normal and in malignant conditions, as well as the therapeutic implications of the interplay between its diverse cellular components and malignant cells.
Collapse
Affiliation(s)
- Valentina Granata
- IRCCS Humanitas Research Hospital, Milan, Italy
- Milan Unit, CNR-IRGB, Milan, Italy
| | - Laura Crisafulli
- IRCCS Humanitas Research Hospital, Milan, Italy
- Milan Unit, CNR-IRGB, Milan, Italy
| | - Claudia Nastasi
- Laboratory of Cancer Pharmacology, Department of Oncology, IRCCS Mario Negri Pharmacological Research Institute, Milan, Italy
| | - Francesca Ficara
- IRCCS Humanitas Research Hospital, Milan, Italy
- Milan Unit, CNR-IRGB, Milan, Italy
| | - Cristina Sobacchi
- IRCCS Humanitas Research Hospital, Milan, Italy
- Milan Unit, CNR-IRGB, Milan, Italy
- *Correspondence: Cristina Sobacchi,
| |
Collapse
|
20
|
Autophagy in mesenchymal progenitors protects mice against bone marrow failure after severe intermittent stress. Blood 2022; 139:690-703. [PMID: 34657154 PMCID: PMC8814682 DOI: 10.1182/blood.2021011775] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 10/01/2021] [Indexed: 11/26/2022] Open
Abstract
The cellular mechanisms required to ensure homeostasis of the hematopoietic niche and the ability of this niche to support hematopoiesis upon stress remain elusive. We here identify Wnt5a in Osterix+ mesenchymal progenitor and stem cells (MSPCs) as a critical factor for niche-dependent hematopoiesis. Mice lacking Wnt5a in MSPCs suffer from stress-related bone marrow (BM) failure and increased mortality. Niche cells devoid of Wnt5a show defective actin stress fiber orientation due to an elevated activity of the small GTPase CDC42. This results in incorrect positioning of autophagosomes and lysosomes, thus reducing autophagy and increasing oxidative stress. In MSPCs from patients from BM failure states which share features of peripheral cytopenia and hypocellular BM, we find similar defects in actin stress fiber orientation, reduced and incorrect colocalization of autophagosomes and lysosomes, and CDC42 activation. Strikingly, a short pharmacological intervention to attenuate elevated CDC42 activation in vivo in mice prevents defective actin-anchored autophagy in MSPCs, salvages hematopoiesis and protects against lethal cytopenia upon stress. In summary, our study identifies Wnt5a as a restriction factor for niche homeostasis by affecting CDC42-regulated actin stress-fiber orientation and autophagy upon stress. Our data further imply a critical role for autophagy in MSPCs for adequate support of hematopoiesis by the niche upon stress and in human diseases characterized by peripheral cytopenias and hypocellular BM.
Collapse
|
21
|
Jann JC, Mossner M, Riabov V, Altrock E, Schmitt N, Flach J, Xu Q, Nowak V, Obländer J, Palme I, Weimer N, Streuer A, Jawhar A, Darwich A, Jawhar M, Metzgeroth G, Nolte F, Hofmann WK, Nowak D. Bone marrow derived stromal cells from myelodysplastic syndromes are altered but not clonally mutated in vivo. Nat Commun 2021; 12:6170. [PMID: 34697318 PMCID: PMC8546146 DOI: 10.1038/s41467-021-26424-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 10/06/2021] [Indexed: 11/15/2022] Open
Abstract
The bone marrow (BM) stroma in myeloid neoplasms is altered and it is hypothesized that this cell compartment may also harbor clonal somatically acquired mutations. By exome sequencing of in vitro expanded mesenchymal stromal cells (MSCs) from n = 98 patients with myelodysplastic syndrome (MDS) and n = 28 healthy controls we show that these cells accumulate recurrent mutations in genes such as ZFX (n = 8/98), RANK (n = 5/98), and others. MDS derived MSCs display higher mutational burdens, increased replicative stress, senescence, inflammatory gene expression, and distinct mutational signatures as compared to healthy MSCs. However, validation experiments in serial culture passages, chronological BM aspirations and backtracking of high confidence mutations by re-sequencing primary sorted MDS MSCs indicate that the discovered mutations are secondary to in vitro expansion but not present in primary BM. Thus, we here report that there is no evidence for clonal mutations in the BM stroma of MDS patients. Bone marrow-derived mesenchymal stroma cells (MSCs) in myeloid neoplasia have been hypothesized to carry somatic mutations and contribute to pathogenesis. Here the authors analyse ex-vivo cultures and primary MSCs derived from patients with myelodysplastic syndromes, finding functional alterations but no evidence of clonal mutations.
Collapse
Affiliation(s)
- Johann-Christoph Jann
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Maximilian Mossner
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Vladimir Riabov
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Eva Altrock
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Nanni Schmitt
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Johanna Flach
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Qingyu Xu
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Verena Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Julia Obländer
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Iris Palme
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Nadine Weimer
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Alexander Streuer
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Ahmed Jawhar
- Department of Orthopedic Surgery, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Ali Darwich
- Department of Orthopedic Surgery, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Mohammad Jawhar
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Georgia Metzgeroth
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Florian Nolte
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Daniel Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany.
| |
Collapse
|
22
|
Emerging Bone Marrow Microenvironment-Driven Mechanisms of Drug Resistance in Acute Myeloid Leukemia: Tangle or Chance? Cancers (Basel) 2021; 13:cancers13215319. [PMID: 34771483 PMCID: PMC8582363 DOI: 10.3390/cancers13215319] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Despite high rates of remission obtained with conventional chemotherapy, the persistence of leukemic cells after treatments, eventually exiting in disease relapse, remains the main challenge in acute myeloid leukemia (AML). Increasing evidence indicates that, besides AML cell mutations, stromal and immune cells, as leukemic microenvironment components, may protect AML cells from therapies. Here, we will recapitulate emerging bone marrow (BM) microenvironment-dependent mechanisms of therapy resistance. The understanding of these processes will help find new drug combinations and conceive novel and more effective treatments. Abstract Acute myeloid leukemia (AML) has been considered for a long time exclusively driven by critical mutations in hematopoietic stem cells. Recently, the contribution of further players, such as stromal and immune bone marrow (BM) microenvironment components, to AML onset and progression has been pointed out. In particular, mesenchymal stromal cells (MSCs) steadily remodel the leukemic niche, not only favoring leukemic cell growth and development but also tuning their responsiveness to treatments. The list of mechanisms driven by MSCs to promote a leukemia drug-resistant phenotype has progressively expanded. Moreover, the relative proportion and the activation status of immune cells in the BM leukemic microenvironment may vary by influencing their reactivity against leukemic cells. In that, the capacity of the stroma to re-program immune cells, thus promoting and/or hampering therapeutic efficacy, is emerging as a crucial aspect in AML biology, adding an extra layer of complexity. Current treatments for AML have mainly focused on eradicating leukemia cells, with little consideration for the leukemia-damaged BM niche. Increasing evidence on the contribution of stromal and immune cells in response to therapy underscores the need to hold the mutual interplay, which takes place in the BM. A careful dissection of these interactions will help provide novel applications for drugs already under experimentation and open a wide array of opportunities for new drug discovery.
Collapse
|
23
|
Balaian E, Wobus M, Bornhäuser M, Chavakis T, Sockel K. Myelodysplastic Syndromes and Metabolism. Int J Mol Sci 2021; 22:ijms222011250. [PMID: 34681910 PMCID: PMC8541058 DOI: 10.3390/ijms222011250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/06/2021] [Accepted: 10/14/2021] [Indexed: 12/01/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are acquired clonal stem cell disorders exhibiting ineffective hematopoiesis, dysplastic cell morphology in the bone marrow, and peripheral cytopenia at early stages; while advanced stages carry a high risk for transformation into acute myeloid leukemia (AML). Genetic alterations are integral to the pathogenesis of MDS. However, it remains unclear how these genetic changes in hematopoietic stem and progenitor cells (HSPCs) occur, and how they confer an expansion advantage to the clones carrying them. Recently, inflammatory processes and changes in cellular metabolism of HSPCs and the surrounding bone marrow microenvironment have been associated with an age-related dysfunction of HSPCs and the emergence of genetic aberrations related to clonal hematopoiesis of indeterminate potential (CHIP). The present review highlights the involvement of metabolic and inflammatory pathways in the regulation of HSPC and niche cell function in MDS in comparison to healthy state and discusses how such pathways may be amenable to therapeutic interventions.
Collapse
Affiliation(s)
- Ekaterina Balaian
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (M.W.); (M.B.)
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence: (E.B.); (K.S.)
| | - Manja Wobus
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (M.W.); (M.B.)
| | - Martin Bornhäuser
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (M.W.); (M.B.)
- National Center for Tumor Diseases, Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Triantafyllos Chavakis
- National Center for Tumor Diseases, Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus Dresden, 01307 Dresden, Germany
| | - Katja Sockel
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (M.W.); (M.B.)
- Correspondence: (E.B.); (K.S.)
| |
Collapse
|
24
|
Lee P, Yim R, Yung Y, Chu HT, Yip PK, Gill H. Molecular Targeted Therapy and Immunotherapy for Myelodysplastic Syndrome. Int J Mol Sci 2021; 22:10232. [PMID: 34638574 PMCID: PMC8508686 DOI: 10.3390/ijms221910232] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/22/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a heterogeneous, clonal hematological disorder characterized by ineffective hematopoiesis, cytopenia, morphologic dysplasia, and predisposition to acute myeloid leukemia (AML). Stem cell genomic instability, microenvironmental aberrations, and somatic mutations contribute to leukemic transformation. The hypomethylating agents (HMAs), azacitidine and decitabine are the standard of care for patients with higher-risk MDS. Although these agents induce responses in up to 40-60% of patients, primary or secondary drug resistance is relatively common. To improve the treatment outcome, combinational therapies comprising HMA with targeted therapy or immunotherapy are being evaluated and are under continuous development. This review provides a comprehensive update of the molecular pathogenesis and immune-dysregulations involved in MDS, mechanisms of resistance to HMA, and strategies to overcome HMA resistance.
Collapse
Affiliation(s)
| | | | | | | | | | - Harinder Gill
- Division of Haematology, Medical Oncology and Haemopoietic Stem Cell Transplantation, Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (P.L.); (R.Y.); (Y.Y.); (H.-T.C.); (P.-K.Y.)
| |
Collapse
|
25
|
Mian SA, Bonnet D. Nature or Nurture? Role of the Bone Marrow Microenvironment in the Genesis and Maintenance of Myelodysplastic Syndromes. Cancers (Basel) 2021; 13:4116. [PMID: 34439269 PMCID: PMC8394536 DOI: 10.3390/cancers13164116] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/18/2022] Open
Abstract
Myelodysplastic syndrome (MDS) are clonal haematopoietic stem cell (HSC) disorders driven by a complex combination(s) of changes within the genome that result in heterogeneity in both clinical phenotype and disease outcomes. MDS is among the most common of the haematological cancers and its incidence markedly increases with age. Currently available treatments have limited success, with <5% of patients undergoing allogeneic HSC transplantation, a procedure that offers the only possible cure. Critical contributions of the bone marrow microenvironment to the MDS have recently been investigated. Although the better understanding of the underlying biology, particularly genetics of haematopoietic stem cells, has led to better disease and risk classification; however, the role that the bone marrow microenvironment plays in the development of MDS remains largely unclear. This review provides a comprehensive overview of the latest developments in understanding the aetiology of MDS, particularly focussing on understanding how HSCs and the surrounding immune/non-immune bone marrow niche interacts together.
Collapse
Affiliation(s)
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London NW1 1AT, UK;
| |
Collapse
|
26
|
Fröbel J, Landspersky T, Percin G, Schreck C, Rahmig S, Ori A, Nowak D, Essers M, Waskow C, Oostendorp RAJ. The Hematopoietic Bone Marrow Niche Ecosystem. Front Cell Dev Biol 2021; 9:705410. [PMID: 34368155 PMCID: PMC8339972 DOI: 10.3389/fcell.2021.705410] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
The bone marrow (BM) microenvironment, also called the BM niche, is essential for the maintenance of fully functional blood cell formation (hematopoiesis) throughout life. Under physiologic conditions the niche protects hematopoietic stem cells (HSCs) from sustained or overstimulation. Acute or chronic stress deregulates hematopoiesis and some of these alterations occur indirectly via the niche. Effects on niche cells include skewing of its cellular composition, specific localization and molecular signals that differentially regulate the function of HSCs and their progeny. Importantly, while acute insults display only transient effects, repeated or chronic insults lead to sustained alterations of the niche, resulting in HSC deregulation. We here describe how changes in BM niche composition (ecosystem) and structure (remodeling) modulate activation of HSCs in situ. Current knowledge has revealed that upon chronic stimulation, BM remodeling is more extensive and otherwise quiescent HSCs may be lost due to diminished cellular maintenance processes, such as autophagy, ER stress response, and DNA repair. Features of aging in the BM ecology may be the consequence of intermittent stress responses, ultimately resulting in the degeneration of the supportive stem cell microenvironment. Both chronic stress and aging impair the functionality of HSCs and increase the overall susceptibility to development of diseases, including malignant transformation. To understand functional degeneration, an important prerequisite is to define distinguishing features of unperturbed niche homeostasis in different settings. A unique setting in this respect is xenotransplantation, in which human cells depend on niche factors produced by other species, some of which we will review. These insights should help to assess deviations from the steady state to actively protect and improve recovery of the niche ecosystem in situ to optimally sustain healthy hematopoiesis in experimental and clinical settings.
Collapse
Affiliation(s)
- Julia Fröbel
- Immunology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Theresa Landspersky
- School of Medicine, Department of Internal Medicine III, Technical University of Munich, Munich, Germany
| | - Gülce Percin
- Immunology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Christina Schreck
- School of Medicine, Department of Internal Medicine III, Technical University of Munich, Munich, Germany
| | - Susann Rahmig
- Immunology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Alessandro Ori
- Proteomics of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Daniel Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marieke Essers
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.,Division Inflammatory Stress in Stem Cells, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Claudia Waskow
- Immunology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany.,Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany.,Department of Medicine III, Technical University Dresden, Dresden, Germany
| | - Robert A J Oostendorp
- School of Medicine, Department of Internal Medicine III, Technical University of Munich, Munich, Germany
| |
Collapse
|