1
|
Ma D, Liang R, Luo Q, Song G. Pressure loading regulates the stemness of liver cancer stem cells via YAP/BMF signaling axis. J Cell Physiol 2025; 240:e31451. [PMID: 39358905 DOI: 10.1002/jcp.31451] [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: 02/08/2024] [Revised: 08/27/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
Cancer stem cells (CSCs) are considered the major cause of the occurrence, progression, chemoresistance/radioresistance, recurrence, and metastasis of cancer. Increased interstitial fluid pressure (IFP) is a key feature of solid tumors. Our previous study showed that the distribution of liver cancer stem cells (LCSCs) correlated with the mechanical heterogeneity within liver cancer tissues. However, the regulation of liver cancer's mechanical microenvironment on the LCSC stemness is not fully understood. Here, we employed a cellular pressure-loading device to investigate the effects of normal IFP (5 mmHg), as well as increased IFP (40 and 200 mmHg) on the stemness of LCSCs. Compared to the control LCSCs (exposure to 5 mmHg pressure loading), the LCSCs exposed to 40 mmHg pressure loading exhibited significantly upregulated expression of CSC markers (CD44, EpCAM, Nanog), enhanced sphere and colony formation capacities, and tumorigenic potential, whereas continuously increased pressure to 200 mmHg suppressed the LCSC characteristics. Mechanistically, pressure loading regulated Yes-associated protein (YAP) activity and Bcl-2 modifying factor (BMF) expression. YAP transcriptionally regulated BMF expression to affect the stemness of LCSCs. Knockdown of YAP and overexpression of BMF attenuated pressure-mediated stemness and tumorgenicity, while YAP-deficient and BMF-deletion recused pressure-dependent stemness on LCSCs, suggesting the involvement of YAP/BMF signaling axis in this process. Together, our findings provide a potential target for overcoming the stemness of CSCs and elucidate the significance of increased IFP in cancer progression.
Collapse
Affiliation(s)
- Di Ma
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Rui Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
2
|
Yu M, Nah GSS, Krishnan V, Sulaimi FNB, Ng KP, Wang C, Bhatt S, Chuah C, Bergstrom DE, Ong ST. The BIM deletion polymorphism potentiates the survival of leukemia stem and progenitor cells and impairs response to targeted therapies. Leukemia 2025; 39:134-143. [PMID: 39438588 DOI: 10.1038/s41375-024-02418-0] [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: 11/27/2023] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
One sixth of human cancers harbor pathogenic germline variants, but few studies have established their functional contribution to cancer outcomes. Here, we developed a humanized mouse model harboring a common East Asian polymorphism, the BIM deletion polymorphism (BDP), which confers resistance to oncogenic kinase inhibitors through generation of non-apoptotic splice isoforms. However, despite its clear role in mediating bulk resistance in patients, the BDP's role in cancer stem and progenitor cells, which initiate disease and possess altered BCL-2 rheostats compared to differentiated tumor cells, remains unknown. To study the role of the BDP in leukemia initiation, we crossed the BDP mouse into a chronic myeloid leukemia (CML) model. We found that the BDP greatly enhanced the fitness of CML cells with a three-fold greater competitive advantage, leading to more aggressive disease. The BDP conferred almost complete resistance to cell death induced by imatinib in CML stem and progenitor cells (LSPCs). Using BH3 profiling, we identified a novel therapeutic vulnerability of BDP LSPCs to MCL-1 antagonists, which we confirmed in primary human LSPCs, and in vivo. Our findings demonstrate the impact of human polymorphisms on the survival of LSPCs and highlight their potential as companion diagnostics for tailored therapies.
Collapse
MESH Headings
- Animals
- Bcl-2-Like Protein 11/genetics
- Mice
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/drug effects
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality
- Polymorphism, Genetic
- Drug Resistance, Neoplasm/genetics
- Imatinib Mesylate/pharmacology
- Imatinib Mesylate/therapeutic use
- Apoptosis
- Cell Survival/genetics
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Molecular Targeted Therapy
Collapse
Affiliation(s)
- Mengge Yu
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Giselle Sek Suan Nah
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Vaidehi Krishnan
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | | | - King Pan Ng
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
- KK Women's and Children's Hospital, Singapore, 229899, Singapore
| | - Chuqi Wang
- Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Shruti Bhatt
- Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Charles Chuah
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
- Department of Haematology, Singapore General Hospital, Singapore, 169608, Singapore
| | | | - S Tiong Ong
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.
- Department of Haematology, Singapore General Hospital, Singapore, 169608, Singapore.
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC, 27708, USA.
| |
Collapse
|
3
|
Li YR, Fang Y, Niu S, Chen Y, Lyu Z, Yang L. Managing allorejection in off-the-shelf CAR-engineered cell therapies. Mol Ther 2024:S1525-0016(24)00762-7. [PMID: 39600090 DOI: 10.1016/j.ymthe.2024.11.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/25/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
Chimeric antigen receptor (CAR)-engineered T (CAR-T) cell therapy has revolutionized the treatment of various diseases, including cancers and autoimmune disorders. However, all US Food and Drug Administration (FDA)-approved CAR-T cell therapies are autologous, and their widespread clinical application is limited by several challenges, such as complex individualized manufacturing, high costs, and the need for patient-specific selection. Allogeneic off-the-shelf CAR-engineered cell therapy offers promising potential due to its immediate availability, consistent quality, potency, and scalability in manufacturing. Nonetheless, significant challenges, including the risks of graft-versus-host disease (GvHD) and host-cell-mediated allorejection, must be addressed. Strategies such as knocking out endogenous T cell receptors (TCRs) or using alternative therapeutic cells with low GvHD risk have shown promise in clinical trials aimed at reducing GvHD. However, mitigating allorejection remains critical for ensuring the long-term sustainability and efficacy of off-the-shelf cell products. In this review, we discuss the immunological basis of allorejection in CAR-engineered therapies and explore various strategies to overcome this challenge. We also highlight key insights from recent clinical trials, particularly related to the sustainability and immunogenicity of allogeneic CAR-engineered cell products, and address manufacturing considerations aimed at minimizing allorejection and optimizing the efficacy of this emerging therapeutic approach.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Ying Fang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Siyue Niu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yuning Chen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zibai Lyu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
4
|
Moiani A, Letort G, Lizot S, Chalumeau A, Foray C, Felix T, Le Clerre D, Temburni-Blake S, Hong P, Leduc S, Pinard N, Marechal A, Seclen E, Boyne A, Mayer L, Hong R, Pulicani S, Galetto R, Gouble A, Cavazzana M, Juillerat A, Miccio A, Duclert A, Duchateau P, Valton J. Non-viral DNA delivery and TALEN editing correct the sickle cell mutation in hematopoietic stem cells. Nat Commun 2024; 15:4965. [PMID: 38862518 PMCID: PMC11166989 DOI: 10.1038/s41467-024-49353-3] [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: 08/08/2023] [Accepted: 06/03/2024] [Indexed: 06/13/2024] Open
Abstract
Sickle cell disease is a devastating blood disorder that originates from a single point mutation in the HBB gene coding for hemoglobin. Here, we develop a GMP-compatible TALEN-mediated gene editing process enabling efficient HBB correction via a DNA repair template while minimizing risks associated with HBB inactivation. Comparing viral versus non-viral DNA repair template delivery in hematopoietic stem and progenitor cells in vitro, both strategies achieve comparable HBB correction and result in over 50% expression of normal adult hemoglobin in red blood cells without inducing β-thalassemic phenotype. In an immunodeficient female mouse model, transplanted cells edited with the non-viral strategy exhibit higher engraftment and gene correction levels compared to those edited with the viral strategy. Transcriptomic analysis reveals that non-viral DNA repair template delivery mitigates P53-mediated toxicity and preserves high levels of long-term hematopoietic stem cells. This work paves the way for TALEN-based autologous gene therapy for sickle cell disease.
Collapse
Affiliation(s)
| | - Gil Letort
- Cellectis S.A., 8 Rue de la Croix Jarry, Paris, France
| | - Sabrina Lizot
- Cellectis S.A., 8 Rue de la Croix Jarry, Paris, France
| | - Anne Chalumeau
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Chloe Foray
- Cellectis S.A., 8 Rue de la Croix Jarry, Paris, France
| | - Tristan Felix
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | | | | | - Patrick Hong
- Cellectis Inc., 430 East 29th Street, New York, NY, USA
| | - Sophie Leduc
- Cellectis S.A., 8 Rue de la Croix Jarry, Paris, France
| | - Noemie Pinard
- Cellectis S.A., 8 Rue de la Croix Jarry, Paris, France
| | - Alan Marechal
- Cellectis S.A., 8 Rue de la Croix Jarry, Paris, France
| | | | - Alex Boyne
- Cellectis Inc., 430 East 29th Street, New York, NY, USA
| | - Louisa Mayer
- Cellectis Inc., 430 East 29th Street, New York, NY, USA
| | - Robert Hong
- Cellectis Inc., 430 East 29th Street, New York, NY, USA
| | | | - Roman Galetto
- Cellectis S.A., 8 Rue de la Croix Jarry, Paris, France
| | - Agnès Gouble
- Cellectis S.A., 8 Rue de la Croix Jarry, Paris, France
| | - Marina Cavazzana
- Biotherapy Clinical Investigation Center, Necker Children's Hospital, Assistance Publique Hopitaux de Paris, Paris, France
- Human Lymphohematopoiesis Laboratory, Imagine Institute, INSERM UMR1163, Paris Cité University, Paris, France
- Biotherapy Department, Necker Children's Hospital, Assistance Publique Hopitaux de Paris, Paris, France
| | | | - Annarita Miccio
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | | | | | - Julien Valton
- Cellectis S.A., 8 Rue de la Croix Jarry, Paris, France.
| |
Collapse
|
5
|
Zehnle PMA, Wu Y, Koleci N, Bohler S, Erlacher M. Lentivirus-Mediated BCL-X L Overexpression Inhibits Stem Cell Apoptosis during Ex Vivo Expansion and Provides Competitive Advantage Following Xenotransplantation. Int J Mol Sci 2024; 25:4105. [PMID: 38612914 PMCID: PMC11012376 DOI: 10.3390/ijms25074105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Hematopoietic reconstitution after hematopoietic stem cell transplantation (HSCT) is influenced by the number of transplanted cells. However, under certain conditions donor cell counts are limited and impair clinical outcome. Hematopoietic stem and progenitor cell (HSPC) expansion prior to HSCT is a widely used method to achieve higher donor cell counts and minimize transplantation-related risks such as graft failure or delayed engraftment. Still, expansion in a non-physiological environment can trigger cell death mechanisms and hence counteract the desired effect. We have shown earlier that during HSCT a relevant amount of HSPCs were lost due to apoptosis and that cell death inhibition in donor HSPCs improved engraftment in xenotransplantation experiments. Here, we assessed the effect of combined ex vivo expansion and cell death inhibition on HSPC yield and their reconstitution potential in vivo. During expansion with cytokines and the small molecule inhibitor StemRegenin 1, concomitant lentiviral overexpression of antiapoptotic BCL-XL resulted in an increased yield of transduced HSPCs. Importantly, BCL-XL overexpression enhanced the reconstitution potential of HSPCs in xenotransplantation experiments in vivo. In contrast, treatment with caspase and necroptosis inhibitors had no favorable effects on HSPC yields nor on cell viability. We postulate that overexpression of antiapoptotic BCL-XL, both during ex vivo expansion and transplantation, is a promising approach to improve the outcome of HSCT in situations with limited donor cell numbers. However, such apoptosis inhibition needs to be transient to avoid long-term sequelae like leukemia.
Collapse
Affiliation(s)
- Patricia M. A. Zehnle
- Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany
- Division of General Pediatrics, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Ying Wu
- Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Naile Koleci
- Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Sheila Bohler
- Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Miriam Erlacher
- Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
6
|
Choe YH, Sorensen J, Garry DJ, Garry MG. Blastocyst complementation and interspecies chimeras in gene edited pigs. Front Cell Dev Biol 2022; 10:1065536. [PMID: 36568986 PMCID: PMC9773398 DOI: 10.3389/fcell.2022.1065536] [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: 10/09/2022] [Accepted: 11/17/2022] [Indexed: 12/13/2022] Open
Abstract
The only curative therapy for many endstage diseases is allograft organ transplantation. Due to the limited supply of donor organs, relatively few patients are recipients of a transplanted organ. Therefore, new strategies are warranted to address this unmet need. Using gene editing technologies, somatic cell nuclear transfer and human induced pluripotent stem cell technologies, interspecies chimeric organs have been pursued with promising results. In this review, we highlight the overall technical strategy, the successful early results and the hurdles that need to be addressed in order for these approaches to produce a successful organ that could be transplanted in patients with endstage diseases.
Collapse
Affiliation(s)
- Yong-ho Choe
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Jacob Sorensen
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Daniel J. Garry
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
- Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN, United States
| | - Mary G. Garry
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
- Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
7
|
Zehnle PMA, Wu Y, Pommerening H, Erlacher M. Stayin‘ alive: BCL-2 proteins in the hematopoietic system. Exp Hematol 2022; 110:1-12. [PMID: 35315320 DOI: 10.1016/j.exphem.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/04/2022]
|
8
|
Wang L, Hensch NR, Bondra K, Sreenivas P, Zhao XR, Chen J, Moreno Campos R, Baxi K, Vaseva AV, Sunkel BD, Gryder BE, Pomella S, Stanton BZ, Zheng S, Chen EY, Rota R, Khan J, Houghton PJ, Ignatius MS. SNAI2-Mediated Repression of BIM Protects Rhabdomyosarcoma from Ionizing Radiation. Cancer Res 2021; 81:5451-5463. [PMID: 34462275 DOI: 10.1158/0008-5472.can-20-4191] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 07/13/2021] [Accepted: 08/26/2021] [Indexed: 11/16/2022]
Abstract
Ionizing radiation (IR) and chemotherapy are mainstays of treatment for patients with rhabdomyosarcoma, yet the molecular mechanisms that underlie the success or failure of radiotherapy remain unclear. The transcriptional repressor SNAI2 was previously identified as a key regulator of IR sensitivity in normal and malignant stem cells through its repression of the proapoptotic BH3-only gene PUMA/BBC3. Here, we demonstrate a clear correlation between SNAI2 expression levels and radiosensitivity across multiple rhabdomyosarcoma cell lines. Modulating SNAI2 levels in rhabdomyosarcoma cells through its overexpression or knockdown altered radiosensitivity in vitro and in vivo. SNAI2 expression reliably promoted overall cell growth and inhibited mitochondrial apoptosis following exposure to IR, with either variable or minimal effects on differentiation and senescence, respectively. Importantly, SNAI2 knockdown increased expression of the proapoptotic BH3-only gene BIM, and chromatin immunoprecipitation sequencing experiments established that SNAI2 is a direct repressor of BIM/BCL2L11. Because the p53 pathway is nonfunctional in the rhabdomyosarcoma cells used in this study, we have identified a new, p53-independent SNAI2/BIM signaling axis that could potentially predict clinical responses to IR treatment and be exploited to improve rhabdomyosarcoma therapy. SIGNIFICANCE: SNAI2 is identified as a major regulator of radiation-induced apoptosis in rhabdomyosarcoma through previously unknown mechanisms independent of p53.
Collapse
Affiliation(s)
- Long Wang
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Science Center, San Antonio, Texas
| | - Nicole R Hensch
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Science Center, San Antonio, Texas.,Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Kathryn Bondra
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Science Center, San Antonio, Texas
| | - Prethish Sreenivas
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Science Center, San Antonio, Texas.,Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Xiang R Zhao
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Science Center, San Antonio, Texas
| | - Jiangfei Chen
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Science Center, San Antonio, Texas.,School of Environmental Safety and Public Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Rodrigo Moreno Campos
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Science Center, San Antonio, Texas.,Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Kunal Baxi
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Science Center, San Antonio, Texas.,Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Angelina V Vaseva
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Science Center, San Antonio, Texas.,Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Benjamin D Sunkel
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio
| | - Berkley E Gryder
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Silvia Pomella
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Benjamin Z Stanton
- Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio.,Department of Biological Chemistry and Pharmacology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Siyuan Zheng
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Science Center, San Antonio, Texas
| | - Eleanor Y Chen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Rossella Rota
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Peter J Houghton
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Science Center, San Antonio, Texas.,Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Myron S Ignatius
- Greehey Children's Cancer Research Institute (GCCRI), UT Health Science Center, San Antonio, Texas. .,Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas
| |
Collapse
|
9
|
Huang Y, Wang S, Ding X, Wu C, Chen J, Hu Z, Du X, Wang G. Inhibition of S-adenosyl-L-homocysteine hydrolase alleviates alloimmune response by down-regulating CD4 + T-cell activation in a mouse heart transplantation model. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1582. [PMID: 33437781 PMCID: PMC7791210 DOI: 10.21037/atm-20-2899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Background Transmethylation reactions play an important role on lymphocyte activation and function. S-adenosyl-L-homocysteine hydrolase (SAHH) inhibitors prevent the feedback of transmethylation reactions by S-adenosyl-L-homocysteine (SAH) accumulation, a competitive antagonist of S-adenosylmethionine (SAM)-dependent methyltransferases. However, the role of SAH in solid organ transplantation is currently unclear. Methods A murine model of cardiac transplantation (BALB/C to C57B/6) was established to assess allograft survival, histology, and T cell infiltration. The reversible SAHH inhibitor, DZ2002, and irreversible SAHH inhibitor, adenosine dialdehyde (AdOx), were used to assess their immunosuppressive effects in murine cardiac transplantation, compared with mice with DMSO. Results Both SAHH inhibitors prolonged the survival of cardiac allografts and alleviated alloimmune response. Notably, AdOx and DZ2002 both eliminated frequencies of Th1 and Th17 in CD4+ T cells in cardiac transplantation, and reduced the frequency of active CD4+ T cell (CD44+ CD62L−). The irreversible SAHH inhibitor facilitated the differentiation of regulatory T cells (Tregs) and increased Bim expression. Furthermore, both SAHH inhibitors alleviated infiltration of CD4+ T cells in cardiac allografts. Conclusions The SAHH inhibitors (AdOx and DZ2002) alleviates allograft rejection in cardiac transplantation by inhibition of CD4+ T alloimmune response. SAHH inhibitors, especially DZ2002, is a promising complementary therapeutic agent in organ transplantation.
Collapse
Affiliation(s)
- Yajun Huang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sufei Wang
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangchao Ding
- Department of Thoracic Surgery, Hubei Provincial People's Hospital, Wuhan University, Wuhan, China
| | - Chuangyan Wu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiuling Chen
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiwei Hu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinling Du
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guohua Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Bohler S, Afreen S, Fernandez-Orth J, Demmerath EM, Molnar C, Wu Y, Weiss JM, Mittapalli VR, Konstantinidis L, Schmal H, Kunze M, Erlacher M. Inhibition of the anti-apoptotic protein MCL-1 severely suppresses human hematopoiesis. Haematologica 2020; 106:3136-3148. [PMID: 33241675 PMCID: PMC8634190 DOI: 10.3324/haematol.2020.252130] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Indexed: 11/16/2022] Open
Abstract
BH3-mimetics inhibiting anti-apoptotic BCL-2 proteins represent a novel and promising class of antitumor drugs. While the BCL-2 inhibitor venetoclax is already approved by the Food and Drug Administration, BCL-XL and MCL-1 inhibitors are currently in early clinical trials. To predict side effects of therapeutic MCL-1 inhibition on the human hematopoietic system, we used RNA interference and the small molecule inhibitor S63845 on cord blood-derived CD34+ cells. Both approaches resulted in almost complete depletion of human hematopoietic stem and progenitor cells. As a consequence, maturation into the different hematopoietic lineages was severely restricted and CD34+ cells expressing MCL-1 shRNA showed a very limited engraftment potential upon xenotransplantation. In contrast, mature blood cells survived normally in the absence of MCL-1. Combined inhibition of MCL-1 and BCL-XL resulted in synergistic effects with relevant loss of colony-forming hematopoietic stem and progenitor cells already at inhibitor concentrations of 0.1 mM each, indicating “synthetic lethality” of the two BH3- mimetics in the hematopoietic system.
Collapse
Affiliation(s)
- Sheila Bohler
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Germany; Faculty of Biology, University of Freiburg
| | - Sehar Afreen
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg
| | - Juncal Fernandez-Orth
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg
| | - Eva-Maria Demmerath
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg
| | - Christian Molnar
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Germany; Faculty of Biology, University of Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg
| | - Ying Wu
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Germany; Faculty of Biology, University of Freiburg
| | - Julia Miriam Weiss
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg
| | - Venugopal Rao Mittapalli
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg
| | - Lukas Konstantinidis
- Department of Orthopedics and Trauma Surgery, Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg
| | - Hagen Schmal
- Department of Orthopedics and Trauma Surgery, Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg
| | - Mirjam Kunze
- Department of Obstetrics and Gynecology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg
| | - Miriam Erlacher
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), Freiburg, Germany and German Cancer Research Center (DKFZ), Heidelberg
| |
Collapse
|
11
|
Afreen S, Bohler S, Müller A, Demmerath EM, Weiss JM, Jutzi JS, Schachtrup K, Kunze M, Erlacher M. BCL-XL expression is essential for human erythropoiesis and engraftment of hematopoietic stem cells. Cell Death Dis 2020; 11:8. [PMID: 31907357 PMCID: PMC6944703 DOI: 10.1038/s41419-019-2203-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023]
Abstract
The anti-apoptotic BCL-2 proteins (BCL-2, BCL-XL, MCL-1, A1, BCL-W) counteract apoptotic signals emerging during development and under stress conditions, and are thus essential for the survival of every cell. While the “BCL-2 addiction” of different cell types is well described in mouse models, there is only limited information available on the role of different anti-apoptotic BCL-2 proteins in a given human cell type. Here we characterize the role of BCL-XL for survival and function of human hematopoietic cells, with the aim to predict hematological side effects of novel BCL-XL-inhibiting BH3-mimetics and to identify hematological malignancies potentially responsive to such inhibitors. Earlier clinical studies have shown that the combined BCL-2/BCL-XL/BCL-W inhibitor, Navitoclax (ABT-263) induces severe thrombocytopenia caused by direct platelet demise and counteracted by increased megakaryopoiesis. In contrast, murine studies have reported important contribution of BCL-XL to survival of late erythroid cells and megakaryocytes. Using lentiviral knockdown, we show that the roles of BCL-XL for human hematopoietic cells are much more pronounced than expected from murine data and clinical trials. Efficient genetic or chemical BCL-XL inhibition resulted in significant loss of human erythroid cells beginning from very early stages of erythropoiesis, and in a reduction of megakaryocytes. Most importantly, BCL-XL deficient human hematopoietic stem cells and multipotent progenitors were reduced in numbers, and they showed a severely impaired capacity to engraft in mice during xenotransplantation. BCL-XL deficiency was fully compensated by BCL-2 overexpression, however, loss of its antagonist BIM did not result in any rescue of human erythroid or stem and progenitor cells. We thus conclude that novel and specific BCL-XL inhibitors might be efficient to treat malignancies of erythroid or megakaryocytic origin, such as polycythemia vera, acute erythroid leukemia, essential thrombocytosis or acute megakaryocytic leukemia. At the same time, it can be expected that they will have more severe hematological side effects than Navitoclax.
Collapse
Affiliation(s)
- Sehar Afreen
- Faculty of Medicine, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany.,University of Freiburg, Freiburg, Germany
| | - Sheila Bohler
- Faculty of Medicine, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany.,University of Freiburg, Freiburg, Germany
| | - Alexandra Müller
- Faculty of Medicine, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Eva-Maria Demmerath
- Faculty of Medicine, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Julia Miriam Weiss
- Faculty of Medicine, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Jonas Samuel Jutzi
- Faculty of Medicine, Section of Molecular Hematology, Department of Medicine I, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Kristina Schachtrup
- Faculty of Medicine, Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Freiburg, Germany
| | - Mirjam Kunze
- Faculty of Medicine, Department of Obstetrics and Gynecology, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany
| | - Miriam Erlacher
- Faculty of Medicine, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, University of Freiburg, Freiburg, Germany. .,German Cancer Consortium (DKTK), Freiburg, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
12
|
Labi V, Peng S, Klironomos F, Munschauer M, Kastelic N, Chakraborty T, Schoeler K, Derudder E, Martella M, Mastrobuoni G, Hernandez-Miranda LR, Lahmann I, Kocks C, Birchmeier C, Kempa S, Quintanilla-Martinez de Fend L, Landthaler M, Rajewsky N, Rajewsky K. Context-specific regulation of cell survival by a miRNA-controlled BIM rheostat. Genes Dev 2019; 33:1673-1687. [PMID: 31699777 PMCID: PMC6942046 DOI: 10.1101/gad.330134.119] [Citation(s) in RCA: 10] [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: 06/27/2019] [Accepted: 10/02/2019] [Indexed: 12/13/2022]
Abstract
Knockout of the ubiquitously expressed miRNA-17∼92 cluster in mice produces a lethal developmental lung defect, skeletal abnormalities, and blocked B lymphopoiesis. A shared target of miR-17∼92 miRNAs is the pro-apoptotic protein BIM, central to life-death decisions in mammalian cells. To clarify the contribution of miR-17∼92:Bim interactions to the complex miR-17∼92 knockout phenotype, we used a system of conditional mutagenesis of the nine Bim 3' UTR miR-17∼92 seed matches. Blocking miR-17∼92:Bim interactions early in development phenocopied the lethal lung phenotype of miR-17∼92 ablation and generated a skeletal kinky tail. In the hematopoietic system, instead of causing the predicted B cell developmental block, it produced a selective inability of B cells to resist cellular stress; and prevented B and T cell hyperplasia caused by Bim haploinsufficiency. Thus, the interaction of miR-17∼92 with a single target is essential for life, and BIM regulation by miRNAs serves as a rheostat controlling cell survival in specific physiological contexts.
Collapse
Affiliation(s)
- Verena Labi
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Siying Peng
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Filippos Klironomos
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Mathias Munschauer
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Nicolai Kastelic
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Tirtha Chakraborty
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Katia Schoeler
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Emmanuel Derudder
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
- Institute for Biomedical Ageing Research, University of Innsbruck, Innsbruck 6020, Austria
| | - Manuela Martella
- Institute of Pathology and Neuropathology and Comprehensive Cancer Center Tübingen, Eberhard-Karls-University, Tübingen 72076, Germany
| | - Guido Mastrobuoni
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Luis R Hernandez-Miranda
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Ines Lahmann
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Christine Kocks
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Carmen Birchmeier
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Stefan Kempa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | | | - Markus Landthaler
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Nikolaus Rajewsky
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
| | - Klaus Rajewsky
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch 13125, Germany
- Program of Cellular and Molecular Medicine, Children's Hospital, and Immune Disease Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
13
|
Singh PK, Roukounakis A, Weber A, Das KK, Sohm B, Villunger A, Garcia-Saez AJ, Häcker G. Dynein light chain binding determines complex formation and posttranslational stability of the Bcl-2 family members Bmf and Bim. Cell Death Differ 2019; 27:434-450. [PMID: 31189926 DOI: 10.1038/s41418-019-0365-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/17/2019] [Accepted: 05/23/2019] [Indexed: 12/31/2022] Open
Abstract
The BH3-only class of Bcl-2 family proteins triggers mitochondrial apoptosis. Several mechanisms are used to restrain the pro-apoptotic activity of these proteins. Dynein light chain (DYNLL) 1 and 2 has been proposed to negatively regulate the activity of Bim and Bmf, respectively, and the Bim-DYNLL1 interaction leads to the formation of large protein complexes on mitochondria. Here we found that Bim and Bmf interact with both isoforms of DYNLL (DYNLL1 and DYNLL2). DYNLL1/2 not only induced homo-dimerization of Bim and Bmf but also led to the formation of ternary complexes (Bim-DYNLL-Bmf), both in cell-free and in cellular systems. DYNLL-induced oligomerization stabilized Bmf in cultured cells and inhibited its degradation by the ubiquitin-independent 20S proteasome in a cell-free system. Surprisingly, overexpression of wild-type Bmf but not of a DYNLL-binding-deficient mutant induced degradation of endogenous Bim in different cell lines, but both variants sensitized to apoptosis. Mutant Bmf incapable of interacting with anti-apoptotic Bcl-2 proteins and of inducing apoptosis still caused Bim degradation. These results suggest that Bmf overexpression-induced Bim degradation is not due to the displacement of Bim from anti-apoptotic Bcl-2 proteins but a direct consequence of the modulation of Bim-DYNLL association. A peptide derived from the DYNLL-binding domain of Bim also led to the degradation of Bim as well as of its preferred binding partner Mcl-1. Thus DYNLL regulates the mitochondrial pathway of apoptosis by determining the stability of Bmf, Bim, and Mcl-1 proteins.
Collapse
Affiliation(s)
- Prafull Kumar Singh
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center - University of Freiburg, 79104, Freiburg, Germany
| | - Aristomenis Roukounakis
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center - University of Freiburg, 79104, Freiburg, Germany
| | - Arnim Weber
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center - University of Freiburg, 79104, Freiburg, Germany
| | - Kushal Kumar Das
- Interfaculty Institute of Biochemistry (IFIB), University of Tübingen, 72076, Tübingen, Germany
| | - Benedicte Sohm
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, 6020, Innsbruck, Austria.,Laboratoire Interdisciplinaire des Environnements Continentaux UMR 7360 CNRS - Université de Lorraine, Metz, France
| | - Andreas Villunger
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Ana J Garcia-Saez
- Interfaculty Institute of Biochemistry (IFIB), University of Tübingen, 72076, Tübingen, Germany
| | - Georg Häcker
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center - University of Freiburg, 79104, Freiburg, Germany. .,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
14
|
Demmerath EM, Bohler S, Kunze M, Erlacher M. In vitro and in vivo evaluation of possible pro-survival activities of PGE2, EGF, TPO and FLT3L on human hematopoiesis. Haematologica 2018; 104:669-677. [PMID: 30442724 PMCID: PMC6442978 DOI: 10.3324/haematol.2018.191569] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 11/14/2018] [Indexed: 12/16/2022] Open
Abstract
Myelosuppression is a major and frequently dose-limiting side effect of anticancer therapy and is responsible for most treatment-related morbidity and mortality. In addition, repeated cycles of DNA damage and cell death of hematopoietic stem and progenitor cells, followed by compensatory proliferation and selection pressure, lead to genomic instability and pave the way for therapy-related myelodysplastic syndromes and secondary acute myeloid leukemia. Protection of hematopoietic stem and progenitor cells from chemo- and radiotherapy in patients with solid tumors would reduce both immediate complications and long-term sequelae. Epidermal growth factor (EGF) and prostaglandin E2 (PGE2) were reported to prevent chemo- or radiotherapy-induced myelosuppression in mice. We tested both molecules for potentially protective effects on human CD34+ cells in vitro and established a xenograft mouse model to analyze stress resistance and regeneration of human hematopoiesis in vivo. EGF was neither able to protect human stem and progenitor cells in vitro nor to promote hematopoietic regeneration following sublethal irradiation in vivo. PGE2 significantly reduced in vitro apoptotic susceptibility of human CD34+ cells to taxol and etoposide. This could, however, be ascribed to reduced proliferation rather than to a change in apoptosis signaling and BCL-2 protein regulation. Accordingly, 16,16-dimethyl-PGE2 (dmPGE2) did not accelerate regeneration of the human hematopoietic system in vivo. Repeated treatment of sublethally irradiated xenograft mice with known antiapoptotic substances, such as human FLT3L and thrombopoietin (TPO), which suppress transcription of the proapoptotic BCL-2 proteins BIM and BMF, also only marginally promoted human hematopoietic regeneration in vivo.
Collapse
Affiliation(s)
- Eva-Maria Demmerath
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg
| | - Sheila Bohler
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg.,Faculty of Biology, University of Freiburg
| | - Mirjam Kunze
- Department of Obstetrics and Gynecology, University Medical Center of Freiburg
| | - Miriam Erlacher
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg .,German Cancer Consortium (DKTK), Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
15
|
Afreen S, Weiss JM, Strahm B, Erlacher M. Concise Review: Cheating Death for a Better Transplant. Stem Cells 2018; 36:1646-1654. [DOI: 10.1002/stem.2901] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/05/2018] [Accepted: 07/15/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Sehar Afreen
- Faculty of Medicine, Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg; University of Freiburg; Freiburg Germany
- Faculty of Biology; University of Freiburg; Freiburg Germany
| | - Julia Miriam Weiss
- Faculty of Medicine, Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg; University of Freiburg; Freiburg Germany
| | - Brigitte Strahm
- Faculty of Medicine, Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg; University of Freiburg; Freiburg Germany
| | - Miriam Erlacher
- Faculty of Medicine, Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg; University of Freiburg; Freiburg Germany
- German Cancer Consortium (DKTK); Freiburg Germany
- German Cancer Research Center (DKFZ); Heidelberg Germany
| |
Collapse
|
16
|
Kurkewich JL, Boucher A, Klopfenstein N, Baskar R, Kapur R, Dahl R. The mirn23a and mirn23b microrna clusters are necessary for proper hematopoietic progenitor cell production and differentiation. Exp Hematol 2017; 59:14-29. [PMID: 29288704 DOI: 10.1016/j.exphem.2017.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 12/17/2017] [Accepted: 12/19/2017] [Indexed: 12/28/2022]
Abstract
Mice deficient for microRNA (miRNA) cluster mirn23a exhibit increased B lymphopoiesis at the expense of myelopoiesis, whereas hematopoietic stem and progenitor cell (HSPC) populations are unchanged. Mammals possess a paralogous mirn23b gene that can give rise to three mature miRNAs (miR-23b, miR-24-1, and miR-27b) that have identical seed/mRNA-targeting sequences to their mirn23a counterparts. To assess whether compound deletion of mirn23a and mirn23b exacerbates the hematopoietic phenotype observed in mirn23a-/- mice, we generated a compound mirn23a-/-mirn23bfl/fl:Mx1-Cre conditional knockout mouse and assayed hematopoietic development after excision of mirn23b. Loss of both genes in adult bone marrow further skewed HSPC differentiation toward B cells at the expense of myeloid cells, demonstrating a dosage-dependent effect on regulating cell differentiation. Strikingly, double-knockout (DKO) mice had decreased bone marrow cellularity with significantly decreased hematopoietic stem cell and HSPC populations, a phenotype not observed in mice deficient for mirn23a alone. Competitive transplantation assays showed decreased contribution of mirn23a-/-mirn23b-/- HSPCs to hematopoietic lineages at 6 and 12 weeks after transplantation. Defects in the proliferation of mirn23a-/-b-/- HSPCs was not observed; however, DKO cells were more apoptotic compared with both wild-type and mirn23a-/- cells. Together, our data show that complete loss of mirn23a/mirn23b miRNAs results in decreased blood production and affects lineage output in a concentration-dependent manner.
Collapse
Affiliation(s)
- Jeffrey L Kurkewich
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA; Harper Cancer Research Institute, South Bend, IN, USA
| | - Austin Boucher
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA; Harper Cancer Research Institute, South Bend, IN, USA
| | - Nathan Klopfenstein
- Harper Cancer Research Institute, South Bend, IN, USA; Department of Microbiology and Immunology, Indiana University School of Medicine, South Bend, IN, USA
| | - Ramdas Baskar
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Reuben Kapur
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Richard Dahl
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA; Harper Cancer Research Institute, South Bend, IN, USA; Department of Microbiology and Immunology, Indiana University School of Medicine, South Bend, IN, USA.
| |
Collapse
|
17
|
Kollek M, Voigt G, Molnar C, Murad F, Bertele D, Krombholz CF, Bohler S, Labi V, Schiller S, Kunze M, Geley S, Niemeyer CM, Garcia-Saez A, Erlacher M. Transient apoptosis inhibition in donor stem cells improves hematopoietic stem cell transplantation. J Exp Med 2017; 214:2967-2983. [PMID: 28882984 PMCID: PMC5626392 DOI: 10.1084/jem.20161721] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 06/14/2017] [Accepted: 07/17/2017] [Indexed: 02/01/2023] Open
Abstract
During hematopoietic stem cell transplantation, a substantial number of donor cells are lost because of apoptotic cell death. Transplantation-associated apoptosis is mediated mainly by the proapoptotic BCL-2 family proteins BIM and BMF, and their proapoptotic function is conserved between mouse and human stem and progenitor cells. Permanent inhibition of apoptosis in donor cells caused by the loss of these BH3-only proteins improves transplantation outcome, but recipients might be exposed to increased risk of lymphomagenesis or autoimmunity. Here, we address whether transient inhibition of apoptosis can serve as a safe but efficient alternative to improve the outcome of stem cell transplantation. We show that transient apoptosis inhibition by short-term overexpression of prosurvival BCL-XL, known to block BIM and BMF, is not only sufficient to increase the viability of hematopoietic stem and progenitor cells during engraftment but also improves transplantation outcome without signs of adverse pathologies. Hence, this strategy represents a promising and novel therapeutic approach, particularly under conditions of limited donor stem cell availability.
Collapse
Affiliation(s)
- Matthias Kollek
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Gesina Voigt
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Molnar
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Fabronia Murad
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Daniela Bertele
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christopher Felix Krombholz
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sheila Bohler
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Verena Labi
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Schiller
- Freiburg Institute for Advanced Studies, University of Freiburg, Freiburg, Germany
- Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany
| | - Mirjam Kunze
- Department of Obstetrics and Gynecology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stephan Geley
- Division of Molecular Pathophysiology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Charlotte M Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ana Garcia-Saez
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Miriam Erlacher
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Freiburg Institute for Advanced Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
18
|
Xie Y, Koch ML, Zhang X, Hamblen MJ, Godinho FJ, Fujiwara Y, Xie H, Klusmann JH, Orkin SH, Li Z. Reduced Erg Dosage Impairs Survival of Hematopoietic Stem and Progenitor Cells. Stem Cells 2017; 35:1773-1785. [PMID: 28436588 DOI: 10.1002/stem.2627] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 03/21/2017] [Indexed: 11/10/2022]
Abstract
ERG, an ETS family transcription factor frequently overexpressed in human leukemia, has been implicated as a key regulator of hematopoietic stem cells. However, how ERG controls normal hematopoiesis, particularly at the stem and progenitor cell level, and how it contributes to leukemogenesis remain incompletely understood. Using homologous recombination, we generated an Erg knockdown allele (Ergkd ) in which Erg expression can be conditionally restored by Cre recombinase. Ergkd/kd animals die at E10.5-E11.5 due to defects in endothelial and hematopoietic cells, but can be completely rescued by Tie2-Cre-mediated restoration of Erg in these cells. In Ergkd/+ mice, ∼40% reduction in Erg dosage perturbs both fetal liver and bone marrow hematopoiesis by reducing the numbers of Lin- Sca-1+ c-Kit+ (LSK) hematopoietic stem and progenitor cells (HSPCs) and megakaryocytic progenitors. By genetic mosaic analysis, we find that Erg-restored HSPCs outcompete Ergkd/+ HSPCs for contribution to adult hematopoiesis in vivo. This defect is in part due to increased apoptosis of HSPCs with reduced Erg dosage, a phenotype that becomes more drastic during 5-FU-induced stress hematopoiesis. Expression analysis reveals that reduced Erg expression leads to changes in expression of a subset of ERG target genes involved in regulating survival of HSPCs, including increased expression of a pro-apoptotic regulator Bcl2l11 (Bim) and reduced expression of Jun. Collectively, our data demonstrate that ERG controls survival of HSPCs, a property that may be used by leukemic cells. Stem Cells 2017;35:1773-1785.
Collapse
Affiliation(s)
- Ying Xie
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Department of Medicine, Boston, Massachusetts, USA
| | - Mia Lee Koch
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Xin Zhang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Melanie J Hamblen
- Division of Hematology and Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Howard Hughes Medical Institute, Boston, Massachusetts, USA
| | - Frank J Godinho
- Division of Hematology and Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Howard Hughes Medical Institute, Boston, Massachusetts, USA
| | - Yuko Fujiwara
- Division of Hematology and Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Howard Hughes Medical Institute, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Huafeng Xie
- Division of Hematology and Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jan-Henning Klusmann
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Stuart H Orkin
- Division of Hematology and Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Howard Hughes Medical Institute, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Zhe Li
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Department of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Tasdogan A, Kumar S, Allies G, Bausinger J, Beckel F, Hofemeister H, Mulaw M, Madan V, Scharffetter-Kochanek K, Feuring-Buske M, Doehner K, Speit G, Stewart AF, Fehling HJ. DNA Damage-Induced HSPC Malfunction Depends on ROS Accumulation Downstream of IFN-1 Signaling and Bid Mobilization. Cell Stem Cell 2016; 19:752-767. [PMID: 27641306 DOI: 10.1016/j.stem.2016.08.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 06/12/2016] [Accepted: 08/09/2016] [Indexed: 01/02/2023]
Abstract
Mouse mutants with an impaired DNA damage response frequently exhibit a set of remarkably similar defects in the HSPC compartment that are of largely unknown molecular basis. Using Mixed-Lineage-Leukemia-5 (Mll5)-deficient mice as prototypical examples, we have identified a mechanistic pathway linking DNA damage and HSPC malfunction. We show that Mll5 deficiency results in accumulation of DNA damage and reactive oxygen species (ROS) in HSPCs. Reduction of ROS efficiently reverses hematopoietic defects, establishing ROS as a major cause of impaired HSPC function. The Ink4a/Arf locus also contributes to HSPC phenotypes, at least in part via promotion of ROS. Strikingly, toxic ROS levels in Mll5-/- mice are critically dependent on type 1 interferon (IFN-1) signaling, which triggers mitochondrial accumulation of full-length Bid. Genetic inactivation of Bid diminishes ROS levels and reverses HSPC defects in Mll5-/- mice. Overall, therefore, our findings highlight an unexpected IFN-1 > Bid > ROS pathway underlying DNA damage-associated HSPC malfunction.
Collapse
Affiliation(s)
- Alpaslan Tasdogan
- Institute of Immunology, University Hospital, 89081 Ulm, Germany; Department of Dermatology, University Hospital, 89081 Ulm, Germany
| | - Suresh Kumar
- Institute of Immunology, University Hospital, 89081 Ulm, Germany
| | - Gabriele Allies
- Institute of Immunology, University Hospital, 89081 Ulm, Germany
| | - Julia Bausinger
- Institute of Human Genetics, University Hospital, 89081 Ulm, Germany
| | - Franziska Beckel
- Institute of Immunology, University Hospital, 89081 Ulm, Germany
| | - Helmut Hofemeister
- Genomics, BioInnovations Zentrum, Technische Universität, 01307 Dresden, Germany
| | - Medhanie Mulaw
- Institute of Experimental Cancer Research, University Clinics, 89081 Ulm, Germany
| | - Vikas Madan
- Institute of Immunology, University Hospital, 89081 Ulm, Germany
| | | | | | - Konstanze Doehner
- Department of Internal Medicine III, University Hospital, 89081 Ulm, Germany
| | - Günter Speit
- Institute of Human Genetics, University Hospital, 89081 Ulm, Germany
| | - A Francis Stewart
- Genomics, BioInnovations Zentrum, Technische Universität, 01307 Dresden, Germany
| | | |
Collapse
|
20
|
Kollek M, Müller A, Egle A, Erlacher M. Bcl-2 proteins in development, health, and disease of the hematopoietic system. FEBS J 2016; 283:2779-810. [DOI: 10.1111/febs.13683] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 01/29/2016] [Accepted: 02/12/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Matthias Kollek
- Division of Pediatric Hematology and Oncology; Department of Pediatrics and Adolescent Medicine; University Medical Center of Freiburg; Germany
- Faculty of Biology; University of Freiburg; Germany
| | - Alexandra Müller
- Division of Pediatric Hematology and Oncology; Department of Pediatrics and Adolescent Medicine; University Medical Center of Freiburg; Germany
| | - Alexander Egle
- Laboratory for Immunological and Molecular Cancer Research; 3rd Medical Department for Hematology; Paracelsus Private Medical University Hospital; Salzburg Austria
| | - Miriam Erlacher
- Division of Pediatric Hematology and Oncology; Department of Pediatrics and Adolescent Medicine; University Medical Center of Freiburg; Germany
| |
Collapse
|
21
|
Krombholz CF, Aumann K, Kollek M, Bertele D, Fluhr S, Kunze M, Niemeyer CM, Flotho C, Erlacher M. Long-term serial xenotransplantation of juvenile myelomonocytic leukemia recapitulates human disease in Rag2-/-γc-/- mice. Haematologica 2016; 101:597-606. [PMID: 26888021 DOI: 10.3324/haematol.2015.138545] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 02/12/2016] [Indexed: 11/09/2022] Open
Abstract
Juvenile myelomonocytic leukemia is a clonal malignant disease affecting young children. Current cure rates, even with allogeneic hematopoietic stem cell transplantation, are no better than 50%-60%. Pre-clinical research on juvenile myelomonocytic leukemia is urgently needed for the identification of novel therapies but is hampered by the unavailability of culture systems. Here we report a xenotransplantation model that allows long-term in vivo propagation of primary juvenile myelomonocytic leukemia cells. Persistent engraftment of leukemic cells was achieved by intrahepatic injection of 1×10(6) cells into newborn Rag2(-/-)γc(-/-) mice or intravenous injection of 5×10(6) cells into 5-week old mice. Key characteristics of juvenile myelomonocytic leukemia were reproduced, including cachexia and clonal expansion of myelomonocytic progenitor cells that infiltrated bone marrow, spleen, liver and, notably, lung. Xenografted leukemia cells led to reduced survival of recipient mice. The stem cell character of juvenile myelomonocytic leukemia was confirmed by successful serial transplantation that resulted in leukemia cell propagation for more than one year. Independence of exogenous cytokines, low donor cell number and slowly progressing leukemia are advantages of the model, which will serve as an important tool to research the pathophysiology of juvenile myelomonocytic leukemia and test novel pharmaceutical strategies such as DNA methyltransferase inhibition.
Collapse
Affiliation(s)
- Christopher Felix Krombholz
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center, Freiburg, Germany Faculty of Biology, University of Freiburg, Germany
| | - Konrad Aumann
- Department of Pathology, University Medical Center, Freiburg, Germany
| | - Matthias Kollek
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center, Freiburg, Germany Faculty of Biology, University of Freiburg, Germany
| | - Daniela Bertele
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center, Freiburg, Germany
| | - Silvia Fluhr
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center, Freiburg, Germany Hermann Staudinger Graduate School, University of Freiburg, Germany
| | - Mirjam Kunze
- Department of Obstetrics and Gynecology, University Medical Center, Freiburg, Germany
| | - Charlotte M Niemeyer
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center, Freiburg, Germany The German Cancer Consortium, Heidelberg, Germany
| | - Christian Flotho
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center, Freiburg, Germany The German Cancer Consortium, Heidelberg, Germany
| | - Miriam Erlacher
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, University Medical Center, Freiburg, Germany The German Cancer Consortium, Heidelberg, Germany
| |
Collapse
|
22
|
Yamashita M, Nitta E, Suda T. Regulation of hematopoietic stem cell integrity through p53 and its related factors. Ann N Y Acad Sci 2015; 1370:45-54. [DOI: 10.1111/nyas.12986] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/11/2015] [Accepted: 11/13/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Masayuki Yamashita
- Department of Cell Differentiation, The Sakaguchi Laboratory of Developmental Biology; School of Medicine, Keio University; Tokyo Japan
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Division of Hematology/Oncology; Department of Medicine, University of California San Francisco; San Francisco California
| | - Eriko Nitta
- Department of Cell Differentiation, The Sakaguchi Laboratory of Developmental Biology; School of Medicine, Keio University; Tokyo Japan
- Department of Cellular and Molecular Medicine, Graduate School of Medicine; Chiba University; Chiba Japan
| | - Toshio Suda
- Department of Cell Differentiation, The Sakaguchi Laboratory of Developmental Biology; School of Medicine, Keio University; Tokyo Japan
- Cancer Science Institute; National University of Singapore; Singapore
- International Research Center for Medical Sciences; Kumamoto University; Kumamoto Japan
| |
Collapse
|
23
|
Antagonism between MCL-1 and PUMA governs stem/progenitor cell survival during hematopoietic recovery from stress. Blood 2015; 125:3273-80. [PMID: 25847014 DOI: 10.1182/blood-2015-01-621250] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/23/2015] [Indexed: 12/11/2022] Open
Abstract
Understanding the critical factors that govern recovery of the hematopoietic system from stress, such as during anticancer therapy and bone marrow transplantation, is of clinical significance. We investigated the importance of the prosurvival proteins myeloid cell leukemia-1 (MCL-1) and B-cell lymphoma-extra large (BCL-XL) in stem/progenitor cell survival and fitness during hematopoietic recovery from stress. Loss of a single Mcl-1 allele, which reduced MCL-1 protein levels, severely compromised hematopoietic recovery from myeloablative challenge and following bone marrow transplantation, whereas BCL-XL was dispensable in both contexts. We identified inhibition of proapoptotic p53 upregulated modulator of apoptosis (PUMA) as the key role of MCL-1 in both settings, with Mcl-1(+/-);Puma(-/-) mice completely protected from the deleterious effects of loss of 1 Mcl-1 allele. These results reveal the molecular mechanisms that govern cell survival during hematopoietic recovery from stress.
Collapse
|
24
|
White MJ, McArthur K, Metcalf D, Lane RM, Cambier JC, Herold MJ, van Delft MF, Bedoui S, Lessene G, Ritchie ME, Huang DCS, Kile BT. Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production. Cell 2015; 159:1549-62. [PMID: 25525874 DOI: 10.1016/j.cell.2014.11.036] [Citation(s) in RCA: 737] [Impact Index Per Article: 73.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 09/22/2014] [Accepted: 11/10/2014] [Indexed: 02/07/2023]
Abstract
Activated caspases are a hallmark of apoptosis induced by the intrinsic pathway, but they are dispensable for cell death and the apoptotic clearance of cells in vivo. This has led to the suggestion that caspases are activated not just to kill but to prevent dying cells from triggering a host immune response. Here, we show that the caspase cascade suppresses type I interferon (IFN) production by cells undergoing Bak/Bax-mediated apoptosis. Bak and Bax trigger the release of mitochondrial DNA. This is recognized by the cGAS/STING-dependent DNA sensing pathway, which initiates IFN production. Activated caspases attenuate this response. Pharmacological caspase inhibition or genetic deletion of caspase-9, Apaf-1, or caspase-3/7 causes dying cells to secrete IFN-β. In vivo, this precipitates an elevation in IFN-β levels and consequent hematopoietic stem cell dysfunction, which is corrected by loss of Bak and Bax. Thus, the apoptotic caspase cascade functions to render mitochondrial apoptosis immunologically silent.
Collapse
Affiliation(s)
- Michael J White
- ACRF Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia.
| | - Kate McArthur
- ACRF Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Donald Metcalf
- Cancer and Haematology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Rachael M Lane
- ACRF Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - John C Cambier
- Integrated Department of Immunology, University of Colorado Denver School of Medicine and National Jewish Health, Denver, CO 80206, USA
| | - Marco J Herold
- Molecular Genetics of Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Mark F van Delft
- Cancer and Haematology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology, The University of Melbourne, Parkville 3010, Australia
| | - Guillaume Lessene
- ACRF Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia; Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville 3010, Australia
| | - Matthew E Ritchie
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia; Department of Mathematics and Statistics, The University of Melbourne, Parkville 3010, Australia
| | - David C S Huang
- Cancer and Haematology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Benjamin T Kile
- ACRF Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia.
| |
Collapse
|
25
|
Izarra A, Moscoso I, Levent E, Cañón S, Cerrada I, Díez-Juan A, Blanca V, Núñez-Gil IJ, Valiente I, Ruíz-Sauri A, Sepúlveda P, Tiburcy M, Zimmermann WH, Bernad A. miR-133a enhances the protective capacity of cardiac progenitors cells after myocardial infarction. Stem Cell Reports 2014; 3:1029-42. [PMID: 25465869 PMCID: PMC4264058 DOI: 10.1016/j.stemcr.2014.10.010] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 10/20/2014] [Accepted: 10/21/2014] [Indexed: 01/06/2023] Open
Abstract
miR-133a and miR-1 are known as muscle-specific microRNAs that are involved in cardiac development and pathophysiology. We have shown that both miR-1 and miR-133a are early and progressively upregulated during in vitro cardiac differentiation of adult cardiac progenitor cells (CPCs), but only miR-133a expression was enhanced under in vitro oxidative stress. miR-1 was demonstrated to favor differentiation of CPCs, whereas miR-133a overexpression protected CPCs against cell death, targeting, among others, the proapoptotic genes Bim and Bmf. miR-133a-CPCs clearly improved cardiac function in a rat myocardial infarction model by reducing fibrosis and hypertrophy and increasing vascularization and cardiomyocyte proliferation. The beneficial effects of miR-133a-CPCs seem to correlate with the upregulated expression of several relevant paracrine factors and the plausible cooperative secretion of miR-133a via exosomal transport. Finally, an in vitro heart muscle model confirmed the antiapoptotic effects of miR-133a-CPCs, favoring the structuration and contractile functionality of the artificial tissue. miR-1 and miR-133a have a role in adult cardiac progenitor cells (CPCs) miR-133a-CPCs protect cardiac function miR-133a-CPCs increase vascularization and protect against hypertrophy miR-133a is enriched in CPCs-derived exosomes
Collapse
Affiliation(s)
- Alberto Izarra
- Immunology and Oncology Department, National Center for Biotechnology, CSIC, 28049 Madrid, Spain; Department of Cardiovascular Development and Repair, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | - Isabel Moscoso
- Department of Cardiovascular Development and Repair, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain; Cardiovascular Area, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15706 Santiago de Compostela, Spain
| | - Elif Levent
- Institute of Pharmacology, Heart Research Center Göttingen, University Medical Center, Georg-August University Göttingen and DZHK (German Center for Cardiovascular Research), 37075 Göttingen, Germany
| | - Susana Cañón
- Immunology and Oncology Department, National Center for Biotechnology, CSIC, 28049 Madrid, Spain; Department of Cardiovascular Development and Repair, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | - Inmaculada Cerrada
- Instituto de Investigación Sanitaria INCLIVA, 46010 Valencia, Spain; Unidad de Cardioregeneración, Hospital La Fe, 46009 Valencia, Spain
| | - Antonio Díez-Juan
- Department of Cardiovascular Development and Repair, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain; Instituto de Investigación Sanitaria INCLIVA, 46010 Valencia, Spain
| | - Vanessa Blanca
- Department of Cardiovascular Development and Repair, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | - Iván-J Núñez-Gil
- Servicio de Cardiología, Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Iñigo Valiente
- Department of Cardiovascular Development and Repair, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | - Amparo Ruíz-Sauri
- Departamento de Patología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain
| | - Pilar Sepúlveda
- Instituto de Investigación Sanitaria INCLIVA, 46010 Valencia, Spain
| | - Malte Tiburcy
- Institute of Pharmacology, Heart Research Center Göttingen, University Medical Center, Georg-August University Göttingen and DZHK (German Center for Cardiovascular Research), 37075 Göttingen, Germany
| | - Wolfram-H Zimmermann
- Institute of Pharmacology, Heart Research Center Göttingen, University Medical Center, Georg-August University Göttingen and DZHK (German Center for Cardiovascular Research), 37075 Göttingen, Germany
| | - Antonio Bernad
- Immunology and Oncology Department, National Center for Biotechnology, CSIC, 28049 Madrid, Spain; Department of Cardiovascular Development and Repair, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
26
|
Carnevalli LS, Scognamiglio R, Cabezas-Wallscheid N, Rahmig S, Laurenti E, Masuda K, Jöckel L, Kuck A, Sujer S, Polykratis A, Erlacher M, Pasparakis M, Essers MAG, Trumpp A. Improved HSC reconstitution and protection from inflammatory stress and chemotherapy in mice lacking granzyme B. ACTA ACUST UNITED AC 2014; 211:769-79. [PMID: 24752302 PMCID: PMC4010905 DOI: 10.1084/jem.20131072] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Granzyme B is expressed by hematopoietic stem cells (HSCs) and stromal cells in response to bacterial products or chemotherapy agents and limits HSC reconstitution potential. The serine protease granzyme B (GzmB) is stored in the granules of cytotoxic T and NK cells and facilitates immune-mediated destruction of virus-infected cells. In this study, we use genetic tools to report novel roles for GzmB as an important regulator of hematopoietic stem cell (HSC) function in response to stress. HSCs lacking the GzmB gene show improved bone marrow (BM) reconstitution associated with increased HSC proliferation and mitochondrial activity. In addition, recipients deficient in GzmB support superior engraftment of wild-type HSCs compared with hosts with normal BM niches. Stimulation of mice with lipopolysaccharide strongly induced GzmB protein expression in HSCs, which was mediated by the TLR4–TRIF–p65 NF-κB pathway. This is associated with increased cell death and GzmB secretion into the BM environment, suggesting an extracellular role of GzmB in modulating HSC niches. Moreover, treatment with the chemotherapeutic agent 5-fluorouracil (5-FU) also induces GzmB production in HSCs. In this situation GzmB is not secreted, but instead causes cell-autonomous apoptosis. Accordingly, GzmB-deficient mice are more resistant to serial 5-FU treatments. Collectively, these results identify GzmB as a negative regulator of HSC function that is induced by stress and chemotherapy in both HSCs and their niches. Blockade of GzmB production may help to improve hematopoiesis in various situations of BM stress.
Collapse
Affiliation(s)
- Larissa S Carnevalli
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Deregulated cell death and lymphocyte homeostasis cause premature lethality in mice lacking the BH3-only proteins Bim and Bmf. Blood 2014; 123:2652-62. [PMID: 24632712 DOI: 10.1182/blood-2013-11-537217] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
BH3 domain-only proteins (BH3-only) proteins are members of the Bcl-2 family that play crucial roles in embryogenesis and the maintenance of tissue homeostasis by triggering apoptotic cell death. The BH3-only protein Bim is critical for developmental apoptosis of lymphocytes, securing establishment of tolerance and for the termination of immune responses. Bim is believed to act in concert with other BH3-only proteins or members of the tumor necrosis factor receptor family in getting rid of unwanted cells. Bmf, a related BH3-only protein, was shown to play a role in B-cell homeostasis and to mediate cell death in response to certain apoptotic triggers, including glucocorticoid, histone deacetylase inhibitors, and overexpression of the c-Myc proto-oncogene. Here we show that Bim and Bmf have overlapping functions during mouse development and coregulate lymphocyte homeostasis and apoptosis in a nonredundant manner. Double deficiency of Bim and Bmf caused more B lymphadenopathy than loss of either BH3-only protein alone, and this was associated with autoimmune glomerulonephritis and a range of malignancies in aged mice. Thus, our results demonstrate that Bim and Bmf act in concert to prevent autoimmunity and malignant disease, strengthening the rational for the development of BH3-only protein mimicking therapeutics for the treatment of such disorders.
Collapse
|
28
|
Yamashita M, Nitta E, Nagamatsu G, Ikushima YM, Hosokawa K, Arai F, Suda T. Nucleostemin is indispensable for the maintenance and genetic stability of hematopoietic stem cells. Biochem Biophys Res Commun 2013; 441:196-201. [DOI: 10.1016/j.bbrc.2013.10.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 10/08/2013] [Indexed: 01/10/2023]
|