1
|
Mašek J, Filipovic I, Van Hul N, Belicová L, Jiroušková M, Oliveira DV, Frontino AM, Hankeova S, He J, Turetti F, Iqbal A, Červenka I, Sarnová L, Verboven E, Brabec T, Björkström NK, Gregor M, Dobeš J, Andersson ER. Jag1 insufficiency alters liver fibrosis via T cell and hepatocyte differentiation defects. EMBO Mol Med 2024; 16:2946-2975. [PMID: 39358604 PMCID: PMC11554675 DOI: 10.1038/s44321-024-00145-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
Fibrosis contributes to tissue repair, but excessive fibrosis disrupts organ function. Alagille syndrome (ALGS, caused by mutations in JAGGED1) results in liver disease and characteristic fibrosis. Here, we show that Jag1Ndr/Ndr mice, a model for ALGS, recapitulate ALGS-like fibrosis. Single-cell RNA-seq and multi-color flow cytometry of the liver revealed immature hepatocytes and paradoxically low intrahepatic T cell infiltration despite cholestasis in Jag1Ndr/Ndr mice. Thymic and splenic regulatory T cells (Tregs) were enriched and Jag1Ndr/Ndr lymphocyte immune and fibrotic capacity was tested with adoptive transfer into Rag1-/- mice, challenged with dextran sulfate sodium (DSS) or bile duct ligation (BDL). Transplanted Jag1Ndr/Ndr lymphocytes were less inflammatory with fewer activated T cells than Jag1+/+ lymphocytes in response to DSS. Cholestasis induced by BDL in Rag1-/- mice with Jag1Ndr/Ndr lymphocytes resulted in periportal Treg accumulation and three-fold less periportal fibrosis than in Rag1-/- mice with Jag1+/+ lymphocytes. Finally, the Jag1Ndr/Ndr hepatocyte expression profile and Treg overrepresentation were corroborated in patients' liver samples. Jag1-dependent hepatic and immune defects thus interact to determine the fibrotic process in ALGS.
Collapse
Affiliation(s)
- Jan Mašek
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden.
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00, Prague 2, Czech Republic.
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, 14183, Sweden.
| | - Iva Filipovic
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Noémi Van Hul
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Lenka Belicová
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Markéta Jiroušková
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
| | - Daniel V Oliveira
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00, Prague 2, Czech Republic
| | - Anna Maria Frontino
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00, Prague 2, Czech Republic
| | - Simona Hankeova
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Jingyan He
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Fabio Turetti
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00, Prague 2, Czech Republic
| | - Afshan Iqbal
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Igor Červenka
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Lenka Sarnová
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
| | - Elisabeth Verboven
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden
| | - Tomáš Brabec
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00, Prague 2, Czech Republic
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
| | - Jan Dobeš
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, 128 00, Prague 2, Czech Republic
| | - Emma R Andersson
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Solna, Stockholm, Sweden.
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, 14183, Sweden.
| |
Collapse
|
2
|
Stankiewicz LN, Salim K, Flaschner EA, Wang YX, Edgar JM, Durland LJ, Lin BZB, Bingham GC, Major MC, Jones RD, Blau HM, Rideout EJ, Levings MK, Zandstra PW, Rossi FMV. Sex-biased human thymic architecture guides T cell development through spatially defined niches. Dev Cell 2024:S1534-5807(24)00539-2. [PMID: 39383865 DOI: 10.1016/j.devcel.2024.09.011] [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: 04/22/2024] [Revised: 06/11/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024]
Abstract
Within the thymus, regulation of the cellular crosstalk directing T cell development depends on spatial interactions within specialized niches. To create a spatially defined map of tissue niches guiding human postnatal T cell development, we employed the multidimensional imaging platform co-detection by indexing (CODEX) as well as cellular indexing of transcriptomes and epitopes sequencing (CITE-seq) and assay for transposase accessible chromatin sequencing (ATAC-seq). We generated age-matched 4- to 5-month-old human postnatal thymus datasets for male and female donors, identifying significant sex differences in both T cell and thymus biology. We demonstrate a possible role for JAG ligands in directing thymic-like dendritic cell development, identify important functions of a population of extracellular matrix (ECM)- fibroblasts, and characterize the medullary niches surrounding Hassall's corpuscles. Together, these data represent an age-matched spatial multiomic resource to investigate how sex-based differences in thymus regulation and T cell development arise, providing an essential resource to understand the mechanisms underlying immune function and dysfunction in males and females.
Collapse
Affiliation(s)
- Laura N Stankiewicz
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Kevin Salim
- Department of Surgery, University of British Columbia, 2775 Laurel Street, Vancouver, BC V5Z 1M9, Canada; BC Children's Hospital Research Institute, 938 W 28th Ave, Vancouver, BC V5Z 4H4, Canada
| | - Emily A Flaschner
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Yu Xin Wang
- Center for Genetic Disorders and Aging, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - John M Edgar
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Lauren J Durland
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Bruce Z B Lin
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Grace C Bingham
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Matthew C Major
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Ross D Jones
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305, USA
| | - Elizabeth J Rideout
- Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Megan K Levings
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada; Department of Surgery, University of British Columbia, 2775 Laurel Street, Vancouver, BC V5Z 1M9, Canada; BC Children's Hospital Research Institute, 938 W 28th Ave, Vancouver, BC V5Z 4H4, Canada
| | - Peter W Zandstra
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T 1Z4, Canada.
| | - Fabio M V Rossi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada; Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 2A1, Canada.
| |
Collapse
|
3
|
Lui PP, Xu JZ, Aziz H, Sen M, Ali N. Jagged-1+ skin Tregs modulate cutaneous wound healing. Sci Rep 2024; 14:20999. [PMID: 39251686 PMCID: PMC11385218 DOI: 10.1038/s41598-024-71512-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/28/2024] [Indexed: 09/11/2024] Open
Abstract
Skin-resident regulatory T cells (Tregs) play an irreplaceable role in orchestrating cutaneous immune homeostasis and repair, including the promotion of hair regeneration via the Notch signaling ligand Jagged-1 (Jag1). While skin Tregs are indispensable for facilitating tissue repair post-wounding, it remains unknown if Jag1-expressing skin Tregs impact wound healing. Using a tamoxifen inducible Foxp3creERT2Jag1fl/fl model, we show that loss of functional Jag1 in Tregs significantly delays the rate of full-thickness wound closure. Unlike in hair regeneration, skin Tregs do not utilize Jag1 to impact epithelial stem cells during wound healing. Instead, mice with Treg-specific Jag1 ablation exhibit a significant reduction in Ly6G + neutrophil accumulation at the wound site. However, during both homeostasis and wound healing, the loss of Jag1 in Tregs does not impact the overall abundance or activation profile of immune cell targets in the skin, such as CD4+ and CD8+ T cells, or pro-inflammatory macrophages. This collectively suggests that skin Tregs may utilize Jag1-Notch signalling to co-ordinate innate cell recruitment under conditions of injury but not homeostasis. Overall, our study demonstrates the importance of Jag1 expression in Tregs to facilitate adequate wound repair in the skin.
Collapse
Affiliation(s)
- Prudence PokWai Lui
- Peter Gorer Department of Immunobiology, King's College London, London, SE1 9RT, UK
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, SE1 9RT, UK
| | - Jessie Z Xu
- Peter Gorer Department of Immunobiology, King's College London, London, SE1 9RT, UK
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, SE1 9RT, UK
| | - Hafsah Aziz
- Peter Gorer Department of Immunobiology, King's College London, London, SE1 9RT, UK
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, SE1 9RT, UK
| | - Monica Sen
- Peter Gorer Department of Immunobiology, King's College London, London, SE1 9RT, UK
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, SE1 9RT, UK
| | - Niwa Ali
- Peter Gorer Department of Immunobiology, King's College London, London, SE1 9RT, UK.
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London, SE1 9RT, UK.
| |
Collapse
|
4
|
Schneider M, Allman A, Maillard I. Regulation of immune cell development, differentiation and function by stromal Notch ligands. Curr Opin Cell Biol 2023; 85:102256. [PMID: 37806295 PMCID: PMC10873072 DOI: 10.1016/j.ceb.2023.102256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/23/2023] [Accepted: 09/10/2023] [Indexed: 10/10/2023]
Abstract
In multicellular organisms, cell-to-cell communication is critical for the regulation of tissue organization. Notch signaling relies on direct interactions between Notch receptors on signal-receiving cells and Notch ligands on adjacent cells. Notch evolved to mediate local cellular interactions that are responsive to spatial cues via dosage-sensitive short-lived signals. Immune cells utilize these unique properties of Notch signaling to direct their development, differentiation, and function. In this review, we explore how immune cells interact through Notch receptors with stromal cells in specialized niches of lymphohematopoietic organs that express Notch-activating ligands. We emphasize factors that control these interactions and focus on how Notch signals communicate spatial, quantitative, and temporal information to program the function of signal-receiving cells in the immune system.
Collapse
Affiliation(s)
- Michael Schneider
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anneka Allman
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ivan Maillard
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Stankiewicz LN, Salim K, Flaschner EA, Wang YX, Edgar JM, Lin BZB, Bingham GC, Major MC, Jones RD, Blau HM, Rideout EJ, Levings MK, Zandstra PW, Rossi FMV. Sex biased human thymic architecture guides T cell development through spatially defined niches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.13.536804. [PMID: 37090676 PMCID: PMC10120731 DOI: 10.1101/2023.04.13.536804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Within the thymus, regulation of the cellular cross-talk directing T cell development is dependent on spatial interactions within specialized niches. To create a holistic, spatially defined map of tissue niches guiding postnatal T cell development we employed the multidimensional imaging platform CO-detection by indEXing (CODEX), as well as CITE-seq and ATAC-seq. We generated age-matched 4-5-month-old postnatal thymus datasets for male and female donors, and identify significant sex differences in both T cell and thymus biology. We demonstrate a crucial role for JAG ligands in directing thymic-like dendritic cell development, reveal important functions of a novel population of ECM- fibroblasts, and characterize the medullary niches surrounding Hassall's corpuscles. Together, these data represent a unique age-matched spatial multiomic resource to investigate how sex-based differences in thymus regulation and T cell development arise, and provide an essential resource to understand the mechanisms underlying immune function and dysfunction in males and females.
Collapse
Affiliation(s)
| | - Kevin Salim
- Department of Surgery, University of British Columbia, Canada
- BC Children’s Hospital Research Institute, Canada
| | - Emily A Flaschner
- School of Biomedical Engineering, University of British Columbia, Canada
| | - Yu Xin Wang
- Center for Genetic Disorders and Aging, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - John M Edgar
- School of Biomedical Engineering, University of British Columbia, Canada
| | - Bruce ZB Lin
- School of Biomedical Engineering, University of British Columbia, Canada
| | - Grace C Bingham
- Department of Biomedical Engineering, University of Virginia, USA
| | - Matthew C Major
- School of Biomedical Engineering, University of British Columbia, Canada
| | - Ross D Jones
- School of Biomedical Engineering, University of British Columbia, Canada
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, USA
| | | | - Megan K Levings
- School of Biomedical Engineering, University of British Columbia, Canada
- Department of Surgery, University of British Columbia, Canada
- BC Children’s Hospital Research Institute, Canada
| | - Peter W Zandstra
- School of Biomedical Engineering, University of British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Canada
- These authors contributed equally
- Lead contact
| | - Fabio MV Rossi
- School of Biomedical Engineering, University of British Columbia, Canada
- These authors contributed equally
- Lead contact
| |
Collapse
|
6
|
Kimura S, Dupee Z, Lima F, Allen R, Kazmi S, Diodati N, Lukacs NW, Kunkel SL, Schaller M. Jagged-1 Reduces Th2 Inflammation and Memory Cell Expansion in Allergic Airway Disease. Immunohorizons 2023; 7:168-176. [PMID: 36729482 PMCID: PMC10563391 DOI: 10.4049/immunohorizons.2300001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 02/03/2023] Open
Abstract
Notch ligands present during interactions between T cells and dendritic cells (DCs) dictate cell phenotype through a myriad of effects including the induction of T cell regulation, survival, and cytokine response. The presence of Notch ligands on DCs varies with the context of the inflammatory response; Jagged-1 is constitutively expressed, whereas Delta-like 1 and Delta-like 4 are induced in response to pathogen exposure. Although Delta-like and Jagged ligands send different signals through the same Notch receptor, the role of these two ligands in peripheral T cell immunity is not clear. The goal of our studies was to determine the role of Jagged-1 in the pathogen-free inflammation induced by OVA during allergic airway disease in mice. Our studies show that a deletion in DC-expressed Jagged-1 causes a significant increase in cytokine production, resulting in increased mucus production and increased eosinophilia in the lungs of mice sensitized and challenged with OVA. We also observed that a reduction of Jagged-1 expression is correlated with increased expression of the Notch 1 receptor on the surface of CD4+ T cells in both the lung and lymph node. Through transfer studies using OT-II transgenic T cells, we demonstrate that Jagged-1 represses the expansion of CD44+CD62L+CCR7+ memory cells and promotes the expansion of CD44+CD62L- effector cells, but it has no effect on the expansion of naive cells during allergic airway disease. These data suggest that Jagged-1 may have different roles in Ag-specific T cell responses, depending on the maturity of the stimulated T cell.
Collapse
Affiliation(s)
- Soichiro Kimura
- Department of Microbiology and Infectious Diseases, Toho University School of Medicine, Tokyo, Japan
- Division of Infection Prevention and Control, Faculty of Pharmaceutical Sciences, Shonan University of Medical Sciences, Kanagawa, Japan
| | - Zadia Dupee
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL; and
| | - Felipe Lima
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL; and
| | - Ronald Allen
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Soha Kazmi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL; and
| | - Nickolas Diodati
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL; and
| | | | | | - Matthew Schaller
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL; and
| |
Collapse
|
7
|
Toribio ML, González-García S. Notch Partners in the Long Journey of T-ALL Pathogenesis. Int J Mol Sci 2023; 24:1383. [PMID: 36674902 PMCID: PMC9866461 DOI: 10.3390/ijms24021383] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological disease that arises from the oncogenic transformation of developing T cells during T-lymphopoiesis. Although T-ALL prognosis has improved markedly in recent years, relapsing and refractory patients with dismal outcomes still represent a major clinical issue. Consequently, understanding the pathological mechanisms that lead to the appearance of this malignancy and developing novel and more effective targeted therapies is an urgent need. Since the discovery in 2004 that a major proportion of T-ALL patients carry activating mutations that turn NOTCH1 into an oncogene, great efforts have been made to decipher the mechanisms underlying constitutive NOTCH1 activation, with the aim of understanding how NOTCH1 dysregulation converts the physiological NOTCH1-dependent T-cell developmental program into a pathological T-cell transformation process. Several molecular players have so far been shown to cooperate with NOTCH1 in this oncogenic process, and different therapeutic strategies have been developed to specifically target NOTCH1-dependent T-ALLs. Here, we comprehensively analyze the molecular bases of the cross-talk between NOTCH1 and cooperating partners critically involved in the generation and/or maintenance and progression of T-ALL and discuss novel opportunities and therapeutic approaches that current knowledge may open for future treatment of T-ALL patients.
Collapse
Affiliation(s)
- María Luisa Toribio
- Immune System Development and Function Unit, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| | | |
Collapse
|
8
|
Ding C, Xu H, Yu Z, Roulis M, Qu R, Zhou J, Oh J, Crawford J, Gao Y, Jackson R, Sefik E, Li S, Wei Z, Skadow M, Yin Z, Ouyang X, Wang L, Zou Q, Su B, Hu W, Flavell RA, Li HB. RNA m 6A demethylase ALKBH5 regulates the development of γδ T cells. Proc Natl Acad Sci U S A 2022; 119:e2203318119. [PMID: 35939687 PMCID: PMC9388086 DOI: 10.1073/pnas.2203318119] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
γδ T cells are an abundant T cell population at the mucosa and are important in providing immune surveillance as well as maintaining tissue homeostasis. However, despite γδ T cells' origin in the thymus, detailed mechanisms regulating γδ T cell development remain poorly understood. N6-methyladenosine (m6A) represents one of the most common posttranscriptional modifications of messenger RNA (mRNA) in mammalian cells, but whether it plays a role in γδ T cell biology is still unclear. Here, we show that depletion of the m6A demethylase ALKBH5 in lymphocytes specifically induces an expansion of γδ T cells, which confers enhanced protection against gastrointestinal Salmonella typhimurium infection. Mechanistically, loss of ALKBH5 favors the development of γδ T cell precursors by increasing the abundance of m6A RNA modification in thymocytes, which further reduces the expression of several target genes including Notch signaling components Jagged1 and Notch2. As a result, impairment of Jagged1/Notch2 signaling contributes to enhanced proliferation and differentiation of γδ T cell precursors, leading to an expanded mature γδ T cell repertoire. Taken together, our results indicate a checkpoint role of ALKBH5 and m6A modification in the regulation of γδ T cell early development.
Collapse
Affiliation(s)
- Chenbo Ding
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
| | - Hao Xu
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Zhibin Yu
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
| | - Manolis Roulis
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Rihao Qu
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
- dProgram of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520
- eDepartment of Pathology, Yale University School of Medicine, New Haven, CT 06510
| | - Jing Zhou
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
| | - Joonseok Oh
- fDepartment of Chemistry, Yale University, New Haven, CT 06520
- gChemical Biology Institute, Yale University, West Haven, CT 06516
| | - Jason Crawford
- fDepartment of Chemistry, Yale University, New Haven, CT 06520
- gChemical Biology Institute, Yale University, West Haven, CT 06516
- hDepartment of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06520
| | - Yimeng Gao
- iSection of Hematology, Yale Cancer Center and Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
- jYale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06520
- kYale RNA Center, Yale University School of Medicine, New Haven, CT 06520
| | - Ruaidhrí Jackson
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Esen Sefik
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Simiao Li
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Zheng Wei
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Mathias Skadow
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Zhinan Yin
- lZhuhai Precision Medical Center, Zhuhai People’s Hospital, Jinan University, Zhuhai 519000, Guangdong, China
- mBiomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China
| | - Xinshou Ouyang
- nSection of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Lei Wang
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qiang Zou
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bing Su
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
| | - Weiguo Hu
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- 2To whom correspondence may be addressed. , , or
| | - Richard A. Flavell
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
- oHHMI, Yale University School of Medicine, New Haven, CT 06520
- 2To whom correspondence may be addressed. , , or
| | - Hua-Bing Li
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
- 2To whom correspondence may be addressed. , , or
| |
Collapse
|
9
|
Multiple Mechanisms of NOTCH1 Activation in Chronic Lymphocytic Leukemia: NOTCH1 Mutations and Beyond. Cancers (Basel) 2022; 14:cancers14122997. [PMID: 35740661 PMCID: PMC9221163 DOI: 10.3390/cancers14122997] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 11/20/2022] Open
Abstract
Simple Summary Mutations of the NOTCH1 gene are a validated prognostic marker in chronic lymphocytic leukemia and a potential predictive marker for anti-CD20-based therapies. At present, the most frequent pathological alteration of the NOTCH1 gene is due to somatic genetic mutations, which have a multifaceted functional impact. However, beside NOTCH1 mutations, other factors may lead to activation of the NOTCH1 pathway, and these include mutations of FBXW7, MED12, SPEN, SF3B1 as well as other B-cell pathways. Understanding the preferential strategies though which CLL cells hijack NOTCH1 signaling may present important clues for designing targeted treatment strategies for the management of CLL. Abstract The Notch signaling pathway plays a fundamental role for the terminal differentiation of multiple cell types, including B and T lymphocytes. The Notch receptors are transmembrane proteins that, upon ligand engagement, undergo multiple processing steps that ultimately release their intracytoplasmic portion. The activated protein ultimately operates as a nuclear transcriptional co-factor, whose stability is finely regulated. The Notch pathway has gained growing attention in chronic lymphocytic leukemia (CLL) because of the high rate of somatic mutations of the NOTCH1 gene. In CLL, NOTCH1 mutations represent a validated prognostic marker and a potential predictive marker for anti-CD20-based therapies, as pathological alterations of the Notch pathway can provide significant growth and survival advantage to neoplastic clone. However, beside NOTCH1 mutation, other events have been demonstrated to perturb the Notch pathway, namely somatic mutations of upstream, or even apparently unrelated, proteins such as FBXW7, MED12, SPEN, SF3B1, as well as physiological signals from other pathways such as the B-cell receptor. Here we review these mechanisms of activation of the NOTCH1 pathway in the context of CLL; the resulting picture highlights how multiple different mechanisms, that might occur under specific genomic, phenotypic and microenvironmental contexts, ultimately result in the same search for proliferative and survival advantages (through activation of MYC), as well as immune escape and therapy evasion (from anti-CD20 biological therapies). Understanding the preferential strategies through which CLL cells hijack NOTCH1 signaling may present important clues for designing targeted treatment strategies for the management of CLL.
Collapse
|
10
|
Valero-Pacheco N, Beaulieu AM. Transcriptional Regulation of Mouse Tissue-Resident Natural Killer Cell Development. Front Immunol 2020; 11:309. [PMID: 32161593 PMCID: PMC7052387 DOI: 10.3389/fimmu.2020.00309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/07/2020] [Indexed: 12/11/2022] Open
Abstract
Natural killer (NK) cells are cytotoxic innate lymphocytes that are well-known for their ability to kill infected or malignant cells. Beyond their roles in tumor surveillance and anti-pathogen defense, more recent studies have highlighted key roles for NK cells in a broad range of biological processes, including metabolic homeostasis, immunomodulation of T cells, contact hypersensitivity, and pregnancy. Consistent with the breadth and diversity of these functions, it is now appreciated that NK cells are a heterogeneous population, comprised of specialized and sometimes tissue-specific subsets with distinct phenotypes and effector functions. Indeed, in addition to the conventional NK cells (cNKs) that are abundant and have been well-studied in the blood and spleen, distinct subsets of tissue-resident NK cells (trNKs) and "helper" Group 1 innate lymphoid cells (ILC1s) have now been described in multiple organs and tissues, including the liver, uterus, thymus, adipose tissue, and skin, among others. The cNK, trNK, and/or helper ILC1 populations that co-exist in these various tissues exhibit both common and distinct developmental requirements, suggesting that a combination of lineage-, subset-, and tissue-specific differentiation processes may contribute to the unique functional properties of these various populations. Here, we provide an overview of the transcriptional regulatory pathways known to instruct the development and differentiation of cNK, trNK, and helper ILC1 populations in specific tissues in mice.
Collapse
Affiliation(s)
- Nuriban Valero-Pacheco
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers – The State University of New Jersey, Newark, NJ, United States
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers – The State University of New Jersey, Newark, NJ, United States
| | - Aimee M. Beaulieu
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers – The State University of New Jersey, Newark, NJ, United States
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers – The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
11
|
Tikhonova AN, Dolgalev I, Hu H, Sivaraj KK, Hoxha E, Cuesta-Domínguez Á, Pinho S, Akhmetzyanova I, Gao J, Witkowski M, Guillamot M, Gutkin MC, Zhang Y, Marier C, Diefenbach C, Kousteni S, Heguy A, Zhong H, Fooksman DR, Butler JM, Economides A, Frenette PS, Adams RH, Satija R, Tsirigos A, Aifantis I. The bone marrow microenvironment at single-cell resolution. Nature 2019; 569:222-228. [PMID: 30971824 DOI: 10.1038/s41586-019-1104-8] [Citation(s) in RCA: 570] [Impact Index Per Article: 114.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/13/2019] [Indexed: 01/06/2023]
Abstract
The bone marrow microenvironment has a key role in regulating haematopoiesis, but its molecular complexity and response to stress are incompletely understood. Here we map the transcriptional landscape of mouse bone marrow vascular, perivascular and osteoblast cell populations at single-cell resolution, both at homeostasis and under conditions of stress-induced haematopoiesis. This analysis revealed previously unappreciated levels of cellular heterogeneity within the bone marrow niche and resolved cellular sources of pro-haematopoietic growth factors, chemokines and membrane-bound ligands. Our studies demonstrate a considerable transcriptional remodelling of niche elements under stress conditions, including an adipocytic skewing of perivascular cells. Among the stress-induced changes, we observed that vascular Notch delta-like ligands (encoded by Dll1 and Dll4) were downregulated. In the absence of vascular Dll4, haematopoietic stem cells prematurely induced a myeloid transcriptional program. These findings refine our understanding of the cellular architecture of the bone marrow niche, reveal a dynamic and heterogeneous molecular landscape that is highly sensitive to stress and illustrate the utility of single-cell transcriptomic data in evaluating the regulation of haematopoiesis by discrete niche populations.
Collapse
Affiliation(s)
- Anastasia N Tikhonova
- Department of Pathology, NYU School of Medicine, New York, NY, USA. .,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA.
| | - Igor Dolgalev
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA.,Applied Bioinformatics Laboratories, NYU School of Medicine, New York, NY, USA
| | - Hai Hu
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Kishor K Sivaraj
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Münster, Germany
| | - Edlira Hoxha
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Álvaro Cuesta-Domínguez
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Sandra Pinho
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, NY, USA
| | | | - Jie Gao
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Matthew Witkowski
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Maria Guillamot
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Michael C Gutkin
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ, USA
| | - Yutong Zhang
- Genome Technology Center, Division of Advanced Research Technologies, NYU School of Medicine, New York, NY, USA
| | - Christian Marier
- Genome Technology Center, Division of Advanced Research Technologies, NYU School of Medicine, New York, NY, USA
| | - Catherine Diefenbach
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Stavroula Kousteni
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Adriana Heguy
- Department of Pathology, NYU School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA.,Genome Technology Center, Division of Advanced Research Technologies, NYU School of Medicine, New York, NY, USA
| | - Hua Zhong
- Division of Biostatistics, Department of Population Health, NYU School of Medicine, New York, NY, USA
| | - David R Fooksman
- Department of Pathology, Albert Einstein College of Medicine, New York, NY, USA
| | - Jason M Butler
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ, USA
| | | | - Paul S Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, New York, NY, USA
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, Münster, Germany
| | | | - Aristotelis Tsirigos
- Department of Pathology, NYU School of Medicine, New York, NY, USA. .,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA. .,Applied Bioinformatics Laboratories, NYU School of Medicine, New York, NY, USA.
| | - Iannis Aifantis
- Department of Pathology, NYU School of Medicine, New York, NY, USA. .,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA.
| |
Collapse
|
12
|
Kamalakar A, Oh MS, Stephenson YC, Ballestas-Naissir SA, Davis ME, Willett NJ, Drissi HM, Goudy SL. A non-canonical JAGGED1 signal to JAK2 mediates osteoblast commitment in cranial neural crest cells. Cell Signal 2018; 54:130-138. [PMID: 30529759 DOI: 10.1016/j.cellsig.2018.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 12/24/2022]
Abstract
During craniofacial development, cranial neural crest (CNC) cells migrate into the developing face and form bone through intramembranous ossification. Loss of JAGGED1 (JAG1) signaling in the CNC cells is associated with maxillary hypoplasia or maxillary bone deficiency (MBD) in mice and recapitulates the MBD seen in humans with Alagille syndrome. JAGGED1, a membrane-bound NOTCH ligand, is required for normal craniofacial development, and Jagged1 mutations in humans are known to cause Alagille Syndrome, which is associated with cardiac, biliary, and bone phenotypes and these children experience increased bony fractures. Previously, we demonstrated deficient maxillary osteogenesis in Wnt1-cre;Jagged1f/f (Jag1CKO) mice by conditional deletion of Jagged1 in maxillary CNC cells. In this study, we investigated the JAG1 signaling pathways in a CNC cell line. Treatment with JAG1 induced osteoblast differentiation and maturation markers, Runx2 and Ocn, respectively, Alkaline Phosphatase (ALP) production, as well as classic NOTCH1 targets, Hes1 and Hey1. While JAG1-induced Hes1 and Hey1 expression levels were predictably decreased after DAPT (NOTCH inhibitor) treatment, JAG1-induced Runx2 and Ocn levels were surprisingly constant in the presence of DAPT, indicating that JAG1 effects in the CNC cells are independent of the canonical NOTCH pathway. JAG1 treatment of CNC cells increased Janus Kinase 2 (JAK2) phosphorylation, which was refractory to DAPT treatment, highlighting the importance of the non-canonical NOTCH pathway during CNC cells osteoblast commitment. Pharmacologic inhibition of JAK2 phosphorylation, with and without DAPT treatment, upon JAG1 induction reduced ALP production and, Runx2 and Ocn gene expression. Collectively, these data suggest that JAK2 is an essential component downstream of a non-canonical JAG1-NOTCH1 pathway through which JAG1 stimulates expression of osteoblast-specific gene targets in CNC cells that contribute to osteoblast differentiation and bone mineralization.
Collapse
Affiliation(s)
| | - Melissa S Oh
- Department of Otolaryngology, Emory University, Atlanta, GA, USA.
| | | | | | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech College of Engineering, Atlanta, GA, USA.
| | - Nick J Willett
- Department of Orthopaedics, Emory University, Atlanta, GA, USA; The Atlanta Veterans Affairs Medical Center, Atlanta, GA, USA.
| | - Hicham M Drissi
- Department of Cell biology, Emory University, Atlanta, GA, USA; Department of Orthopaedics, Emory University, Atlanta, GA, USA; The Atlanta Veterans Affairs Medical Center, Atlanta, GA, USA.
| | - Steven L Goudy
- Department of Otolaryngology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
13
|
Brillantes M, Beaulieu AM. Transcriptional control of natural killer cell differentiation. Immunology 2018; 156:111-119. [PMID: 30450565 DOI: 10.1111/imm.13017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/24/2018] [Accepted: 10/24/2018] [Indexed: 01/01/2023] Open
Abstract
Natural killer (NK) cells are highly specialized cytotoxic lymphocytes that provide protection against pathogens and malignant cells. They develop from common lymphoid progenitors via a multi-stage lineage commitment and differentiation process that gives rise to mature NK cells with potent cytotoxic functionality. Although generally considered cells of the innate immune system, recent studies have demonstrated that NK cells have the capacity to mount immune responses with features of adaptive immunity, including robust antigen-specific clonal-like expansion and the generation of long-lived memory cells that mediate enhanced recall responses. Here, we discuss specific transcription factors that have been shown to commonly and uniquely regulate NK cell development and effector and memory responses in experimental mouse models.
Collapse
Affiliation(s)
- Marc Brillantes
- Rutgers Graduate School of Biomedical Sciences, Rutgers - The State University of New Jersey, Newark, NJ, USA
| | - Aimee M Beaulieu
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers - The State University of New Jersey, Newark, NJ, USA.,Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers - The State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
14
|
Colombo M, Mirandola L, Chiriva-Internati M, Basile A, Locati M, Lesma E, Chiaramonte R, Platonova N. Cancer Cells Exploit Notch Signaling to Redefine a Supportive Cytokine Milieu. Front Immunol 2018; 9:1823. [PMID: 30154786 PMCID: PMC6102368 DOI: 10.3389/fimmu.2018.01823] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022] Open
Abstract
Notch signaling is a well-known key player in the communication between adjacent cells during organ development, when it controls several processes involved in cell differentiation. Notch-mediated communication may occur through the interaction of Notch receptors with ligands on adjacent cells or by a paracrine/endocrine fashion, through soluble molecules that can mediate the communication between cells at distant sites. Dysregulation of Notch pathway causes a number of disorders, including cancer. Notch hyperactivation may be caused by mutations of Notch-related genes, dysregulated upstream pathways, or microenvironment signals. Cancer cells may exploit this aberrant signaling to "educate" the surrounding microenvironment cells toward a pro-tumoral behavior. This may occur because of key cytokines secreted by tumor cells or it may involve the microenvironment through the activation of Notch signaling in stromal cells, an event mediated by a direct cell-to-cell contact and resulting in the increased secretion of several pro-tumorigenic cytokines. Up to now, review articles were mainly focused on Notch contribution in a specific tumor context or immune cell populations. Here, we provide a comprehensive overview on the outcomes of Notch-mediated pathological interactions in different tumor settings and on the molecular and cellular mediators involved in this process. We describe how Notch dysregulation in cancer may alter the cytokine network and its outcomes on tumor progression and antitumor immune response.
Collapse
Affiliation(s)
- Michela Colombo
- Department of Health Sciences, Università degli Studi di Milano, Milano, Italy
| | | | - Maurizio Chiriva-Internati
- Kiromic Biopharma Inc., Houston, TX, United States.,Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andrea Basile
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy
| | - Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Milano, Italy.,Humanitas Clinical and Research Center, Rozzano, Italy
| | - Elena Lesma
- Department of Health Sciences, Università degli Studi di Milano, Milano, Italy
| | | | - Natalia Platonova
- Department of Health Sciences, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
15
|
García-León MJ, Fuentes P, de la Pompa JL, Toribio ML. Dynamic regulation of NOTCH1 activation and Notch ligand expression in human thymus development. Development 2018; 145:dev.165597. [PMID: 30042180 DOI: 10.1242/dev.165597] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/06/2018] [Indexed: 01/22/2023]
Abstract
T-cell development is a complex dynamic process that relies on ordered stromal signals delivered to thymus-seeding progenitors that migrate throughout different thymus microenvironments (TMEs). Particularly, Notch signaling provided by thymic epithelial cells (TECs) is crucial for T-cell fate specification and generation of mature T cells. Four canonical Notch ligands (Dll1, Dll4, Jag1 and Jag2) are expressed in the thymus, but their spatial distribution in functional TMEs is largely unknown, especially in humans, and their impact on Notch1 activation during T-lymphopoiesis remains undefined. Based on immunohistochemistry and quantitative confocal microscopy of fetal, postnatal and adult human and mouse thymus samples, we show that spatial regulation of Notch ligand expression defines discrete Notch signaling niches and dynamic species-specific TMEs. We further show that Notch ligand expression, particularly DLL4, is tightly regulated in cortical TECs during human thymus ontogeny and involution. Also, we provide the first evidence that NOTCH1 activation is induced in vivo in CD34+ progenitors and developing thymocytes at particular cortical niches of the human fetal and postnatal thymus. Collectively, our results show that human thymopoiesis involves complex spatiotemporal regulation of Notch ligand expression, which ensures the coordinated delivery of niche-specific NOTCH1 signals required for dynamic T-cell development.
Collapse
Affiliation(s)
- María J García-León
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo de Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| | - Patricia Fuentes
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo de Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain.,CIBER CV, 28029 Madrid, Spain
| | - María L Toribio
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo de Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| |
Collapse
|
16
|
Stage-specific roles for Zmiz1 in Notch-dependent steps of early T-cell development. Blood 2018; 132:1279-1292. [PMID: 30076146 DOI: 10.1182/blood-2018-02-835850] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/29/2018] [Indexed: 12/15/2022] Open
Abstract
Notch1 signaling must elevate to high levels in order to drive the proliferation of CD4-CD8- double-negative (DN) thymocytes and progression to the CD4+CD8+ double-positive (DP) stage through β-selection. During this critical phase of pre-T-cell development, which is also known as the DN-DP transition, it is unclear whether the Notch1 transcriptional complex strengthens its signal output as a discrete unit or through cofactors. We previously showed that the protein inhibitor of activated STAT-like coactivator Zmiz1 is a context-dependent cofactor of Notch1 in T-cell leukemia. We also showed that withdrawal of Zmiz1 generated an early T-lineage progenitor (ETP) defect. Here, we show that this early defect seems inconsistent with loss-of-Notch1 function. In contrast, at the later pre-T-cell stage, withdrawal of Zmiz1 impaired the DN-DP transition by inhibiting proliferation, like withdrawal of Notch. In pre-T cells, but not ETPs, Zmiz1 cooperatively regulated Notch1 target genes Hes1, Lef1, and Myc. Enforced expression of either activated Notch1 or Myc partially rescued the Zmiz1-deficient DN-DP defect. We identified residues in the tetratricopeptide repeat (TPR) domain of Zmiz1 that bind Notch1. Mutating only a single residue impaired the Zmiz1-Notch1 interaction, Myc induction, the DN-DP transition, and leukemic proliferation. Similar effects were seen using a dominant-negative TPR protein. Our studies identify stage-specific roles of Zmiz1. Zmiz1 is a context-specific cofactor for Notch1 during Notch/Myc-dependent thymocyte proliferation, whether normal or malignant. Finally, we highlight a vulnerability in leukemic cells that originated from a developmentally important Zmiz1-Notch1 interaction that is hijacked during transformation from normal pre-T cells.
Collapse
|
17
|
Abstract
Notch drives critical decisions in a multitude of developmental decisions in many invertebrate and vertebrate organisms including flies, worms, fish, mice and humans. Therefore, it is not surprising that Notch family members also play a key role in cell fate choices in the vertebrate immune system. This review highlights the critical function of Notch in the development of mature T lymphocytes from hematopoietic precursors and describes the role of Notch in mature T cell activation, proliferation and differentiation.
Collapse
|
18
|
Low S, Barnes JL, Zammit PS, Beauchamp JR. Delta-Like 4 Activates Notch 3 to Regulate Self-Renewal in Skeletal Muscle Stem Cells. Stem Cells 2017; 36:458-466. [PMID: 29230914 DOI: 10.1002/stem.2757] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 11/10/2017] [Accepted: 11/19/2017] [Indexed: 12/31/2022]
Abstract
Notch signaling is essential to maintain skeletal muscle stem cells in quiescence. However, the precise roles of different Notch receptors are incompletely defined. Here, we demonstrate a role for Notch3 (N3) in the self-renewal of muscle stem cells. We found that N3 is active in quiescent C2C12 reserve cells (RCs), and N3 over-expression and knockdown studies in C2C12 and primary satellite cells reveal a role in self-renewal. The Notch ligand Delta-like 4 (Dll4) is expressed by newly formed myotubes and interaction with this ligand is sufficient to maintain N3 activity in quiescent C2C12 RCs to prevent activation and progression into the cell cycle. Thus, our data suggest a model whereby during regeneration, expression of Dll4 by nascent muscle fibers triggers N3 signaling in associated muscle stem cells to recruit them to quiescence, thereby renewing the stem cell pool. Stem Cells 2018;36:458-466.
Collapse
Affiliation(s)
- SiewHui Low
- Carnegie Institution for Science, Baltimore, Maryland, USA.,Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, Surrey, United Kingdom
| | - Josephine L Barnes
- Centre for Inflammation and Tissue Repair, University College London, London, United Kingdom
| | - Peter S Zammit
- Randall Centre for Cellular and Molecular Biophysics, King's College London, London, United Kingdom
| | - Jonathan R Beauchamp
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, Surrey, United Kingdom
| |
Collapse
|
19
|
Sanghez V, Luzzi A, Clarke D, Kee D, Beuder S, Rux D, Osawa M, Madrenas J, Chou TF, Kyba M, Iacovino M. Notch activation is required for downregulation of HoxA3-dependent endothelial cell phenotype during blood formation. PLoS One 2017; 12:e0186818. [PMID: 29073173 PMCID: PMC5658089 DOI: 10.1371/journal.pone.0186818] [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: 06/09/2017] [Accepted: 10/09/2017] [Indexed: 01/02/2023] Open
Abstract
Hemogenic endothelium (HE) undergoes endothelial-to-hematopoietic transition (EHT) to generate blood, a process that requires progressive down-regulation of endothelial genes and induction of hematopoietic ones. Previously, we have shown that the transcription factor HoxA3 prevents blood formation by inhibiting Runx1 expression, maintaining endothelial gene expression and thus blocking EHT. In the present study, we show that HoxA3 also prevents blood formation by inhibiting Notch pathway. HoxA3 induced upregulation of Jag1 ligand in endothelial cells, which led to cis-inhibition of the Notch pathway, rendering the HE nonresponsive to Notch signals. While Notch activation alone was insufficient to promote blood formation in the presence of HoxA3, activation of Notch or downregulation of Jag1 resulted in a loss of the endothelial phenotype which is a prerequisite for EHT. Taken together, these results demonstrate that Notch pathway activation is necessary to downregulate endothelial markers during EHT.
Collapse
Affiliation(s)
- Valentina Sanghez
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, United States of America.,Los Angeles Biomedical Research Institute, Torrance, CA, United States of America
| | - Anna Luzzi
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, United States of America.,Los Angeles Biomedical Research Institute, Torrance, CA, United States of America
| | - Don Clarke
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, United States of America.,Los Angeles Biomedical Research Institute, Torrance, CA, United States of America
| | - Dustin Kee
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, United States of America.,Los Angeles Biomedical Research Institute, Torrance, CA, United States of America
| | - Steven Beuder
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, United States of America.,Los Angeles Biomedical Research Institute, Torrance, CA, United States of America
| | - Danielle Rux
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States of America
| | - Mitsujiro Osawa
- CiRA
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Joaquín Madrenas
- Los Angeles Biomedical Research Institute, Torrance, CA, United States of America.,Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Tsui-Fen Chou
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, United States of America.,Los Angeles Biomedical Research Institute, Torrance, CA, United States of America
| | - Michael Kyba
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States of America
| | - Michelina Iacovino
- Division of Medical Genetics, Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, United States of America.,Los Angeles Biomedical Research Institute, Torrance, CA, United States of America
| |
Collapse
|
20
|
Dar AA, Bhat SA, Gogoi D, Gokhale A, Chiplunkar SV. Inhibition of Notch signalling has ability to alter the proximal and distal TCR signalling events in human CD3 + αβ T-cells. Mol Immunol 2017; 92:116-124. [PMID: 29078088 DOI: 10.1016/j.molimm.2017.10.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/26/2017] [Accepted: 10/17/2017] [Indexed: 01/13/2023]
Abstract
The Notch signalling pathway is an important regulator of T cell function and is known to regulate the effector functions of T cells driven by T cell receptor (TCR). However, the mechanism integrating these pathways in human CD3+ αβ T cells is not well understood. The present study was carried out to investigate how Notch and TCR driven signalling are synchronized in human αβ T cells. Differential expression of Notch receptors, ligands, and target genes is observed on human αβ T cells which are upregulated on stimulation with α-CD3/CD28 mAb. Inhibition of Notch signalling by GSI-X inhibited the activation of T cells and affected proximal T cell signalling by regulating CD3-ζ chain expression. Inhibition of Notch signalling decreased the protein expression of CD3-ζ chain and induced expression of E3 ubiquitin ligase (GRAIL) in human αβ T cells. Apart from affecting proximal TCR signalling, Notch signalling also regulated the distal TCR signalling events. In the absence of Notch signalling, α-CD3/CD28 mAb induced activation and IFN-γ production by αβ T cells was down-modulated. The absence of Notch signalling in human αβ T cells inhibited proliferative responses despite strong signalling through TCR and IL-2 receptor. This study shows how Notch signalling cooperates with TCR signalling by regulating CD3-ζ chain expression to support proliferation and activation of human αβ T cells.
Collapse
Affiliation(s)
- Asif A Dar
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, Maharashtra 400094, India
| | - Sajad A Bhat
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, Maharashtra 400094, India
| | - Dimpu Gogoi
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, Maharashtra 400094, India
| | - Abhiram Gokhale
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Shubhada V Chiplunkar
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, Maharashtra 400094, India.
| |
Collapse
|
21
|
Martín-Gayo E, González-García S, García-León MJ, Murcia-Ceballos A, Alcain J, García-Peydró M, Allende L, de Andrés B, Gaspar ML, Toribio ML. Spatially restricted JAG1-Notch signaling in human thymus provides suitable DC developmental niches. J Exp Med 2017; 214:3361-3379. [PMID: 28947612 PMCID: PMC5679173 DOI: 10.1084/jem.20161564] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 06/18/2017] [Accepted: 08/08/2017] [Indexed: 01/06/2023] Open
Abstract
Martín-Gayo et al. report that human early thymic progenitors can undergo a GATA2-dependent myeloid developmental program leading to resident dendritic cells (DCs) upon JAG1-Notch activation. The identification of JAG1+ DC-permissive intrathymic niches validates the human thymus as a DC-poietic organ. A key unsolved question regarding the developmental origin of conventional and plasmacytoid dendritic cells (cDCs and pDCs, respectively) resident in the steady-state thymus is whether early thymic progenitors (ETPs) could escape T cell fate constraints imposed normally by a Notch-inductive microenvironment and undergo DC development. By modeling DC generation in bulk and clonal cultures, we show here that Jagged1 (JAG1)-mediated Notch signaling allows human ETPs to undertake a myeloid transcriptional program, resulting in GATA2-dependent generation of CD34+ CD123+ progenitors with restricted pDC, cDC, and monocyte potential, whereas Delta-like1 signaling down-regulates GATA2 and impairs myeloid development. Progressive commitment to the DC lineage also occurs intrathymically, as myeloid-primed CD123+ monocyte/DC and common DC progenitors, equivalent to those previously identified in the bone marrow, are resident in the normal human thymus. The identification of a discrete JAG1+ thymic medullary niche enriched for DC-lineage cells expressing Notch receptors further validates the human thymus as a DC-poietic organ, which provides selective microenvironments permissive for DC development.
Collapse
Affiliation(s)
- Enrique Martín-Gayo
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sara González-García
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - María J García-León
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alba Murcia-Ceballos
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Alcain
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marina García-Peydró
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Luis Allende
- Immunology Department, i+12 Research Institute, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Belén de Andrés
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - María L Gaspar
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - María L Toribio
- Department of Cell Biology and Immunology, Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
22
|
Raevskaya AA, Savvateeva MV, Bukhinnik SS, Kandarakov OF, Butylin PA, Zhuk SV, Demin AM, Krasnov VP, Zaritsky AY, Belyavsky AV. Murine and human hematopoietic progenitor cultures grown on stromal layers expressing Notch ligands. Mol Biol 2017. [DOI: 10.1134/s0026893317020169] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
23
|
Kobayashi M, Nabinger SC, Bai Y, Yoshimoto M, Gao R, Chen S, Yao C, Dong Y, Zhang L, Rodriguez S, Yashiro-Ohtani Y, Pear WS, Carlesso N, Yoder MC, Kapur R, Kaplan MH, Daniel Lacorazza H, Zhang ZY, Liu Y. Protein Tyrosine Phosphatase PRL2 Mediates Notch and Kit Signals in Early T Cell Progenitors. Stem Cells 2017; 35:1053-1064. [PMID: 28009085 DOI: 10.1002/stem.2559] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 11/23/2016] [Accepted: 12/08/2016] [Indexed: 01/18/2023]
Abstract
The molecular pathways regulating lymphoid priming, fate, and development of multipotent bone marrow hematopoietic stem and progenitor cells (HSPCs) that continuously feed thymic progenitors remain largely unknown. While Notch signal is indispensable for T cell specification and differentiation, the downstream effectors are not well understood. PRL2, a protein tyrosine phosphatase that regulates hematopoietic stem cell proliferation and self-renewal, is highly expressed in murine thymocyte progenitors. Here we demonstrate that protein tyrosine phosphatase PRL2 and receptor tyrosine kinase c-Kit are critical downstream targets and effectors of the canonical Notch/RBPJ pathway in early T cell progenitors. While PRL2 deficiency resulted in moderate defects of thymopoiesis in the steady state, de novo generation of T cells from Prl2 null hematopoietic stem cells was significantly reduced following transplantation. Prl2 null HSPCs also showed impaired T cell differentiation in vitro. We found that Notch/RBPJ signaling upregulated PRL2 as well as c-Kit expression in T cell progenitors. Further, PRL2 sustains Notch-mediated c-Kit expression and enhances stem cell factor/c-Kit signaling in T cell progenitors, promoting effective DN1-DN2 transition. Thus, we have identified a critical role for PRL2 phosphatase in mediating Notch and c-Kit signals in early T cell progenitors. Stem Cells 2017;35:1053-1064.
Collapse
Affiliation(s)
| | - Sarah C Nabinger
- Department of Pediatrics, Herman B Wells Center for Pediatric Research
| | - Yunpeng Bai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Momoko Yoshimoto
- Department of Pediatrics, Herman B Wells Center for Pediatric Research
| | - Rui Gao
- Department of Pediatrics, Herman B Wells Center for Pediatric Research
| | - Sisi Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Chonghua Yao
- Department of Pediatrics, Herman B Wells Center for Pediatric Research
| | - Yuanshu Dong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Lujuan Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sonia Rodriguez
- Department of Pediatrics, Herman B Wells Center for Pediatric Research
| | - Yumi Yashiro-Ohtani
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Warren S Pear
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Nadia Carlesso
- Department of Pediatrics, Herman B Wells Center for Pediatric Research
| | - Mervin C Yoder
- Department of Pediatrics, Herman B Wells Center for Pediatric Research
| | - Reuben Kapur
- Department of Pediatrics, Herman B Wells Center for Pediatric Research
| | - Mark H Kaplan
- Department of Pediatrics, Herman B Wells Center for Pediatric Research
| | - Hugo Daniel Lacorazza
- Department of Pathology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA
| | - Yan Liu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
24
|
Yucel D, Kocabas F. Developments in Hematopoietic Stem Cell Expansion and Gene Editing Technologies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1079:103-125. [DOI: 10.1007/5584_2017_114] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
25
|
Brown G, Sanchez-Garcia I. Is lineage decision-making restricted during tumoral reprograming of haematopoietic stem cells? Oncotarget 2016; 6:43326-41. [PMID: 26498146 PMCID: PMC4791235 DOI: 10.18632/oncotarget.6145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 09/29/2015] [Indexed: 01/11/2023] Open
Abstract
Within the past years there have been substantial changes to our understanding of haematopoiesis and cells that initiate and sustain leukemia. Recent studies have revealed that developing haematopoietic stem and progenitor cells are much more heterogeneous and versatile than has been previously thought. This versatility includes cells using more than one route to a fate and cells having progressed some way towards a cell type retaining other lineage options as clandestine. These notions impact substantially on our understanding of the origin and nature of leukemia. An important question is whether leukemia stem cells are as versatile as their cell of origin as an abundance of cells belonging to a lineage is often a feature of overt leukemia. In this regard, we examine the coming of age of the "leukemia stem cell" theory and the notion that leukemia, like normal haematopoiesis, is a hierarchically organized tissue. We examine evidence to support the notion that whilst cells that initiate leukemia have multi-lineage potential, leukemia stem cells are reprogrammed by further oncogenic insults to restrict their lineage decision-making. Accordingly, evolution of a sub-clone of lineage-restricted malignant cells is a key feature of overt leukemia.
Collapse
Affiliation(s)
- Geoffrey Brown
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Isidro Sanchez-Garcia
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Campus M. de Unamuno s/n, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| |
Collapse
|
26
|
Geiger TL, Sun JC. Development and maturation of natural killer cells. Curr Opin Immunol 2016; 39:82-9. [PMID: 26845614 PMCID: PMC4801705 DOI: 10.1016/j.coi.2016.01.007] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/12/2016] [Accepted: 01/14/2016] [Indexed: 12/21/2022]
Abstract
Natural killer (NK) cells are innate lymphocytes that are critical for host protection against pathogens and cancer due to their ability to rapidly release inflammatory cytokines and kill infected or transformed cells. In the 40 years since their initial discovery, much has been learned about how this important cellular lineage develops and functions. We now know that NK cells are the founding members of an expanded family of lymphocyte known as innate lymphoid cells (ILC). Furthermore, we have recently discovered that NK cells can possess features of adaptive immunity such as antigen specificity and long-lived memory responses. Here we will review our current understanding of the molecular mechanisms driving development of NK cells from the common lymphoid progenitor (CLP) to mature NK cells, and from activated effectors to long-lived memory NK cells.
Collapse
Affiliation(s)
- Theresa L Geiger
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, New York, NY 10065, United States; Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Joseph C Sun
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, New York, NY 10065, United States; Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States; Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY 10065, United States.
| |
Collapse
|
27
|
Sukarawan W, Peetiakarawach K, Pavasant P, Osathanon T. Effect of Jagged-1 and Dll-1 on osteogenic differentiation by stem cells from human exfoliated deciduous teeth. Arch Oral Biol 2016; 65:1-8. [PMID: 26826998 DOI: 10.1016/j.archoralbio.2016.01.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 01/18/2016] [Accepted: 01/19/2016] [Indexed: 02/09/2023]
Abstract
OBJECTIVE The aim of the present study was to determine the influence of Notch ligands, Jagged-1 and Dll-1, on osteogenic differentiation by stem cells from human exfoliated deciduous teeth. DESIGN Notch ligands were immobilized on tissue culture surface using an indirect affinity immobilization technique. Cells from the remaining of dental pulp tissues from human deciduous teeth were isolated and characterized using flow cytometry and differentiation assay. Alkaline phosphatase (ALP) enzymatic activity, osteogenic marker gene expression, and mineralization were determined using ALP assay, real-time polymerase chain reaction, and alizarin red staining, respectively. RESULTS The isolated cells exhibited CD44, CD90, and CD105 expression but lack of CD45 expression. Further, these cells were able to differentiate toward osteogenic lineage. The upregulation of HES-1 and HEY-1 was observed in those cells on Dll-1 and Jagged-1 coated surface. The significant increase of ALP activity and mineralization was noted in those cells seeded on Jagged-1 surface and these results were attenuated when cells were pretreated with gamma secretase inhibitor. The significant upregulation of ALP and collagen type I gene expression was also observed in those cells seeded on Jagged-1 surface. The inconsistent Dll-1 induced osteogenic differentiation was found and high Dll-1 immobilized dose (50 nM) slightly enhanced alkaline phosphatase enzymatic activity. However, the statistical significant difference was not obtained as compared to the hFc control. CONCLUSION The surface immobilization of Notch ligands, Jagged-1 and Dll-1, likely to enhance osteogenic differentiation of SHEDs. However, Jagged-1 had more ability in enhancing osteogenic differentiation than Dll-1 in our model.
Collapse
Affiliation(s)
- Waleerat Sukarawan
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330 Thailand; Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330 Thailand.
| | - Karnnapas Peetiakarawach
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330 Thailand; Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330 Thailand
| | - Prasit Pavasant
- Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330 Thailand; Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330 Thailand
| | - Thanaphum Osathanon
- Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330 Thailand; Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330 Thailand.
| |
Collapse
|
28
|
Elemam NM, Mekky RY, El-Ekiaby NM, El Sobky SA, El Din MAM, Esmat G, Abdelaziz AI. Repressing PU.1 by miR-29a∗ in NK cells of HCV patients, diminishes its cytolytic effect on HCV infected cell models. Hum Immunol 2015; 76:687-94. [PMID: 26429314 DOI: 10.1016/j.humimm.2015.09.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 09/24/2015] [Accepted: 09/27/2015] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Natural killer cells are immune safeguards against HCV infection. PU.1 is a pivotal transcription factor in the development of NK cells. This study aimed at studying the regulatory effect of miRNAs on both development and function of NK cells isolated from HCV patients. METHODS NK cells were isolated from 17 chronic HCV patients and 12 healthy controls; after which miRNA and mRNA were quantified using qRT-PCR. Manipulating miRNA expression using mimics and antagomirs, was performed followed by investigating downstream targets as well as viral abundance. RESULTS PU.1 expression levels were upregulated in NK cells of HCV patients. In silico analysis revealed PU.1 to be a potential downstream target of miR-29a(∗), where miR-29a(∗) overexpression in NK cells caused a significant downregulation in PU.1 mRNA. Forcing miR-29a(∗) caused a downregulation of the cytotoxicity determinant NK activating receptor (NKG2D) via upregulation of miR-155. Moreover, perforin-1 mRNA was found to be downregulated upon forcing the expression of miR-29a(∗) in NK cells of HCV patients. This decrease in NK cytolytic function was accompanied by an 80% viral load increase in cocultured HCVcc cell models. CONCLUSIONS This study showed that HCV infection might abrogate NK cytotoxic potential through altering PU.1, NKG2D receptor and perforin molecules.
Collapse
Affiliation(s)
- Noha Mousaad Elemam
- The Molecular Pathology Research Group, Department of Pharmacology and Toxicology, German University in Cairo, New Cairo City, Main Entrance Al Tagamoa Al Khames, 11835 Cairo, Egypt
| | - Radwa Yehia Mekky
- The Molecular Pathology Research Group, Department of Pharmacology and Toxicology, German University in Cairo, New Cairo City, Main Entrance Al Tagamoa Al Khames, 11835 Cairo, Egypt
| | - Nada Magdy El-Ekiaby
- The Molecular Pathology Research Group, Department of Pharmacology and Toxicology, German University in Cairo, New Cairo City, Main Entrance Al Tagamoa Al Khames, 11835 Cairo, Egypt
| | - Shereen Ahmed El Sobky
- The Molecular Pathology Research Group, Department of Pharmaceutical Biology, German University in Cairo, New Cairo City, Main Entrance Al Tagamoa Al Khames, 11835 Cairo, Egypt
| | | | - Gamal Esmat
- Department of Tropical Medicine and Hepatology, Cairo University, Kasr El-Aini St, Cairo, Egypt
| | - Ahmed Ihab Abdelaziz
- The Molecular Pathology Research Group, Department of Pharmacology and Toxicology, German University in Cairo, New Cairo City, Main Entrance Al Tagamoa Al Khames, 11835 Cairo, Egypt.
| |
Collapse
|
29
|
Ogawa M, Ogawa S, Bear CE, Ahmadi S, Chin S, Li B, Grompe M, Keller G, Kamath BM, Ghanekar A. Directed differentiation of cholangiocytes from human pluripotent stem cells. Nat Biotechnol 2015; 33:853-61. [PMID: 26167630 DOI: 10.1038/nbt.3294] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 06/19/2015] [Indexed: 12/28/2022]
Abstract
Although bile duct disorders are well-recognized causes of liver disease, the molecular and cellular events leading to biliary dysfunction are poorly understood. To enable modeling and drug discovery for biliary disease, we describe a protocol that achieves efficient differentiation of biliary epithelial cells (cholangiocytes) from human pluripotent stem cells (hPSCs) through delivery of developmentally relevant cues, including NOTCH signaling. Using three-dimensional culture, the protocol yields cystic and/or ductal structures that express mature biliary markers, including apical sodium-dependent bile acid transporter, secretin receptor, cilia and cystic fibrosis transmembrane conductance regulator (CFTR). We demonstrate that hPSC-derived cholangiocytes possess epithelial functions, including rhodamine efflux and CFTR-mediated fluid secretion. Furthermore, we show that functionally impaired hPSC-derived cholangiocytes from cystic fibrosis patients are rescued by CFTR correctors. These findings demonstrate that mature cholangiocytes can be differentiated from hPSCs and used for studies of biliary development and disease.
Collapse
Affiliation(s)
- Mina Ogawa
- 1] McEwen Centre for Regenerative Medicine, University Health Network, Toronto, Ontario, Canada. [2] Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Shinichiro Ogawa
- McEwen Centre for Regenerative Medicine, University Health Network, Toronto, Ontario, Canada
| | - Christine E Bear
- Program in Molecular Structure &Function, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Saumel Ahmadi
- Program in Molecular Structure &Function, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Stephanie Chin
- Program in Molecular Structure &Function, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Bin Li
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon, USA
| | - Markus Grompe
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon, USA
| | - Gordon Keller
- 1] McEwen Centre for Regenerative Medicine, University Health Network, Toronto, Ontario, Canada. [2] Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. [3] Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Binita M Kamath
- 1] Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Ontario, Canada. [2] Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Anand Ghanekar
- 1] Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada. [2] Division of General Surgery, University Health Network, Toronto, Ontario, Canada. [3] Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Abstract
The lymphocyte family has expanded significantly in recent years to include not only the adaptive lymphocytes (T cells, B cells) and NK cells, but also several additional innate lymphoid cell (ILC) types. ILCs lack clonally distributed antigen receptors characteristic of adaptive lymphocytes and instead respond exclusively to signaling via germline-encoded receptors. ILCs resemble T cells more closely than any other leukocyte lineage at the transcriptome level and express many elements of the core T cell transcriptional program, including Notch, Gata3, Tcf7, and Bcl11b. We present our current understanding of the shared and distinct transcriptional regulatory mechanisms involved in the development of adaptive T lymphocytes and closely related ILCs. We discuss the possibility that a core set of transcriptional regulators common to ILCs and T cells establish enhancers that enable implementation of closely aligned effector pathways. Studies of the transcriptional regulation of lymphopoiesis will support the development of novel therapeutic approaches to correct early lymphoid developmental defects and aberrant lymphocyte function.
Collapse
Affiliation(s)
- Maria Elena De Obaldia
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | | |
Collapse
|
31
|
Abstract
Natural killer (NK) cells are innate lymphocytes that survey the environment and protect the host from infected and cancerous cells. As their name implies, NK cells represent an early line of defense during pathogen invasion by directly killing infected cells and secreting inflammatory cytokines. Although the function of NK cells was first described more than four decades ago, the development of this cytotoxic lineage is not well understood. In recent years, we have begun to identify specific transcription factors that control each stage of development and maturation, from ontogeny of the NK cell progenitor to the effector functions of activated NK cells in peripheral organs. This chapter highlights the transcription factors that are unique to NK cells, or shared between NK cells and other hematopoietic cell lineages, but govern the biology of this cytolytic lymphocyte.
Collapse
Affiliation(s)
- Joseph C Sun
- Memorial Sloan Kettering Cancer Center, Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, 408 East 69th Street, ZRC-1402, New York, NY, 10065, USA.
| |
Collapse
|
32
|
Beaulieu AM, Bezman NA, Lee JE, Matloubian M, Sun JC, Lanier LL. MicroRNA function in NK-cell biology. Immunol Rev 2013; 253:40-52. [PMID: 23550637 DOI: 10.1111/imr.12045] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The important role of microRNAs in directing immune responses has become increasingly clear. Here, we highlight discoveries uncovering the role of specific microRNAs in regulating the development and function of natural killer (NK) cells. Furthermore, we discuss the impact of NK cells on the entire immune system during global and specific microRNA ablation in the settings of inflammation, infection, and immune dysregulation.
Collapse
Affiliation(s)
- Aimee M Beaulieu
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | | |
Collapse
|
33
|
Phelan JD, Saba I, Zeng H, Kosan C, Messer MS, Olsson HA, Fraszczak J, Hildeman DA, Aronow BJ, Möröy T, Grimes HL. Growth factor independent-1 maintains Notch1-dependent transcriptional programming of lymphoid precursors. PLoS Genet 2013; 9:e1003713. [PMID: 24068942 PMCID: PMC3772063 DOI: 10.1371/journal.pgen.1003713] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 06/25/2013] [Indexed: 11/19/2022] Open
Abstract
Growth factor independent 1 (Gfi1) is a transcriptional repressor originally identified as a gene activated in T-cell leukemias induced by Moloney-murine-leukemia virus infection. Notch1 is a transmembrane receptor that is frequently mutated in human T-cell acute lymphoblastic leukemia (T-ALL). Gfi1 is an important factor in the initiation and maintenance of lymphoid leukemias and its deficiency significantly impedes Notch dependent initiation of T-ALL in animal models. Here, we show that immature hematopoietic cells require Gfi1 to competently integrate Notch-activated signaling. Notch1 activation coupled with Gfi1 deficiency early in T-lineage specification leads to a dramatic loss of T-cells, whereas activation in later stages leaves development unaffected. In Gfi1 deficient multipotent precursors, Notch activation induces lethality and is cell autonomous. Further, without Gfi1, multipotent progenitors do not maintain Notch1-activated global expression profiles typical for T-lineage precursors. In agreement with this, we find that both lymphoid-primed multipotent progenitors (LMPP) and early T lineage progenitors (ETP) do not properly form or function in Gfi1−/− mice. These defects correlate with an inability of Gfi1−/− progenitors to activate lymphoid genes, including IL7R, Rag1, Flt3 and Notch1. Our data indicate that Gfi1 is required for hematopoietic precursors to withstand Notch1 activation and to maintain Notch1 dependent transcriptional programming to determine early T-lymphoid lineage identity. Understanding the mechanisms that protect lymphoid cells from transformation is a critical first step in developing therapies against blood cancers. Recently, we demonstrated that the Growth factor independent-1 transcriptional repressor protein is required for cancer development driven by activation of Notch1 signaling. Here, we investigated the mechanisms by which Gfi1 protects lymphoid transformation. Using complex genetic mouse models to delete Gfi1 and activate Notch1, we demonstrate that Gfi1 is required to maintain both the homeostatic levels of Notch1 target genes in normal lymphoid precursors in the bone marrow, as well as to maintain the supraphysiologic levels of Notch1 signaling present in pre-malignant lymphoid progenitors. Consequently, without Gfi1 the pool of premalignant cells available for transformation is depleted. Our data provide additional insight into the multiple mechanisms by which developmental networks may have evolved to protect lymphoid cells from transformation.
Collapse
Affiliation(s)
- James D. Phelan
- Division of Cellular and Molecular Immunology; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Ingrid Saba
- Institut de recherches cliniques de Montréal IRCM, Montréal, Québec, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Hui Zeng
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Christian Kosan
- Institut de recherches cliniques de Montréal IRCM, Montréal, Québec, Canada
| | - Malynda S. Messer
- Division of Cellular and Molecular Immunology; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - H. Andre Olsson
- Division of Cellular and Molecular Immunology; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Jennifer Fraszczak
- Institut de recherches cliniques de Montréal IRCM, Montréal, Québec, Canada
| | - David A. Hildeman
- Division of Cellular and Molecular Immunology; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Bruce J. Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Tarik Möröy
- Institut de recherches cliniques de Montréal IRCM, Montréal, Québec, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Québec, Canada
- * E-mail: (TM); (HLG)
| | - H. Leighton Grimes
- Division of Cellular and Molecular Immunology; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
- Division of Experimental Hematology; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
- * E-mail: (TM); (HLG)
| |
Collapse
|
34
|
GATA-3 promotes T-cell specification by repressing B-cell potential in pro-T cells in mice. Blood 2013; 121:1749-59. [PMID: 23287858 DOI: 10.1182/blood-2012-06-440065] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Transcription factors orchestrate T-lineage differentiation in the thymus. One critical checkpoint involves Notch1 signaling that instructs T-cell commitment at the expense of the B-lineage program. While GATA-3 is required for T-cell specification, its mechanism of action is poorly understood. We show that GATA-3 works in concert with Notch1 to commit thymic progenitors to the T-cell lineage via 2 distinct pathways. First, GATA-3 orchestrates a transcriptional “repertoire” that is required for thymocyte maturation up to and beyond the pro-T-cell stage. Second, GATA-3 critically suppresses a latent B-cell potential in pro–T cells. As such, GATA-3 is essential to sealing in Notch-induced T-cell fate in early thymocyte precursors by promoting T-cell identity through the repression of alternative developmental options.
Collapse
|
35
|
Brown G, Hughes PJ, Ceredig R. The versatile landscape of haematopoiesis: are leukaemia stem cells as versatile? Crit Rev Clin Lab Sci 2012; 49:232-40. [PMID: 23153117 DOI: 10.3109/10408363.2012.742487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Since the early 1980s, developing haematopoietic cells have been categorised into three well-defined compartments: multi-potent haematopoietic stem cells (HSC), which are able to self-renew, followed by haematopoietic progenitor cells (HPC), which undergo decision-making and age as they divide rather than self-renew, and the final compartment of functional blood and immune cells. The classic model of haematopoiesis divides cells into two families, myeloid and lymphoid, and dictates a route to a particular cell fate. New discoveries question these long-held principles, including: (i) the identification of lineage-biased cells that self-renew; (ii) a strict myeloid/lymphoid dichotomy is refuted by the existence of progenitors with lymphoid potential and an incomplete set of myeloid potentials; (iii) there are multiple routes to some end cell types; and (iv) thymocyte progenitor cells that have progressed some way along this pathway retain clandestine myeloid options. In essence, the progeny of HSC are more versatile and the process of haematopoiesis is more flexible than previously thought. Here we examine this new way of viewing haematopoiesis and the impact of rewriting an account of haematopoiesis on our understanding of what goes awry in leukaemia.
Collapse
Affiliation(s)
- Geoffrey Brown
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | | | | |
Collapse
|
36
|
Dervovic DD, Ciofani M, Kianizad K, Zúñiga-Pflücker JC. Comparative and functional evaluation of in vitro generated to ex vivo CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2012; 189:3411-20. [PMID: 22925927 DOI: 10.4049/jimmunol.1200979] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The generation of the cytotoxic CD8 T cell response is dependent on the functional outcomes imposed by the intrathymic constraints of differentiation and self-tolerance. Although thymic function can be partly replicated in vitro using OP9-DL1 cell cultures to yield CD8 αβ TCR-bearing cells from hematopoietic progenitor cells, a comprehensive and functional assessment of entirely in vitro generated CD8 T cells derived from bone marrow hematopoietic stem cells has not been established and remains controversial. In this study, we demonstrate that a phenotypic, molecular, and functional signature of in vitro derived CD8 T cells is akin to that of ex vivo CD8 T cells, although several significant differences were also observed. Transfer of in vitro derived CD8 T cells into syngeneic and immunodeficient host mice showed no graft-versus-host response, whereas a robust homeostatic proliferation was observed, respectively. These findings, along with a diverse and broad TCR repertoire expressed by the in vitro derived CD8 T cells, allowed for the successful generation of Ag-specific T cells to be obtained from an entirely in vitro generated CD8 T cell pool. These findings support the use of Ag-specific in vitro derived effector CD8 T cells for immune reconstitution approaches, which would be amenable to further tailoring for their use against viral infections or malignancies.
Collapse
Affiliation(s)
- Dzana D Dervovic
- Department of Immunology, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| | | | | | | |
Collapse
|
37
|
Li J, Cai H, Jin J, Wang Q, Miao D. X-ray irradiation selectively kills thymocytes of different stages and impairs the maturation of donor-derived CD4(+)CD8(+) thymocytes in recipient thymus. J Biomed Res 2012; 26:355-64. [PMID: 23554771 PMCID: PMC3613732 DOI: 10.7555/jbr.26.20120003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 02/03/2012] [Accepted: 02/25/2012] [Indexed: 12/13/2022] Open
Abstract
The aim of the present study was to determine whether the sensitivity of thymocytes to X-ray radiation depends on their proliferative states and whether radiation impairs the maturation of donor-derived thymocytes in recipient thymus. We assigned 8-week-old C57BL/6J mice into three treatment groups: 1) untreated; 2) X-ray radiation; 3) X-ray radiation plus bone marrow transplantation with donor bone marrow cells from transgenic mice expressing enhanced green fluorescent protein (GFP) on a universal promoter. After 4 weeks, the size of the thymus, the number and proliferation of thymocytes and ratios of different stage thymocytes were analyzed by immunohistochemistry and flow cytometry. The results showed that: 1) CD4+CD8+ thymocytes were more sensitive to X-ray radiation-induced cell death than other thymocytes; 2) the proliferative capacity of CD4+CD8+ thymocytes was higher than that of other thymocytes; 3) the size of the thymus, the number of thymocytes and ratios of thymocytes of different stages in irradiated mice recovered to the normal level of untreated mice by bone marrow transplantation; 4) the ratio of GFP-positive CD4+CD8+ thymocytes increased significantly, whereas the ratio of GFP-positive CD4+ or CD8+ thymocytes decreased significantly. These results indicate that the degree of sensitivity of thymocytes to X-ray radiation depends on their proliferative states and radiation impairs the maturation of donor-derived CD4+CD8+ thymocytes in recipient thymus.
Collapse
Affiliation(s)
- Jinbo Li
- The Research Center for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | | | | | | | | |
Collapse
|
38
|
Abstract
Notch signaling is critical during multiple stages of T cell development in both mouse and human. Evidence has emerged in recent years that this pathway might regulate T-lineage differentiation differently between both species. Here, we review our current understanding of how Notch signaling is activated and used during human T cell development. First, we set the stage by describing the developmental steps that make up human T cell development before describing the expression profiles of Notch receptors, ligands, and target genes during this process. To delineate stage-specific roles for Notch signaling during human T cell development, we subsequently try to interpret the functional Notch studies that have been performed in light of these expression profiles and compare this to its suggested role in the mouse.
Collapse
|
39
|
González-García S, García-Peydró M, Alcain J, Toribio ML. Notch1 and IL-7 receptor signalling in early T-cell development and leukaemia. Curr Top Microbiol Immunol 2012; 360:47-73. [PMID: 22695916 DOI: 10.1007/82_2012_231] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Notch receptors are master regulators of many aspects of development and tissue renewal in metazoans. Notch1 activation is essential for T-cell specification of bone marrow-derived multipotent progenitors that seed the thymus, and for proliferation and further progression of early thymocytes along the T-cell lineage. Deregulated activation of Notch1 significantly contributes to the generation of T-cell acute lymphoblastic leukaemia (T-ALL). In addition to Notch1 signals, survival and proliferation signals provided by the IL-7 receptor (IL-7R) are also required during thymopoiesis. Our understanding of the molecular mechanisms controlling stage-specific survival and proliferation signals provided by Notch1 and IL-7R has recently been improved by the discovery that the IL-7R is a transcriptional target of Notch1. Thus, Notch1 controls T-cell development, in part by regulating the stage- and lineage-specific expression of IL-7R. The finding that induction of IL-7R expression downstream of Notch1 also occurs in T-ALL highlights the important contribution that deregulated IL-7R expression and function may have in this pathology. Confirming this notion, oncogenic IL7R gain-of-function mutations have recently been identified in childhood T-ALL. Here we discuss the fundamental role of Notch1 and IL-7R signalling pathways in physiological and pathological T-cell development in mice and men, highlighting their close molecular underpinnings.
Collapse
Affiliation(s)
- Sara González-García
- Centro de Biología Molecular, Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | | | | | | |
Collapse
|
40
|
Shah DK, Zúñiga-Pflücker JC. Notch receptor-ligand interactions during T cell development, a ligand endocytosis-driven mechanism. Curr Top Microbiol Immunol 2012; 360:19-46. [PMID: 22581027 DOI: 10.1007/82_2012_225] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Notch signaling plays an important role during the development of different cell types and tissues. The role of Notch signaling in lymphocyte development, in particular in the development and commitment to the T cell lineage, has been the focus of research for many years. Notch signaling is absolutely required during the commitment and early stages of T cell development. Activation of the Notch signaling pathway is initiated by ligand-receptor interactions and appears to require active endocytosis of Notch ligands. Studies addressing the mechanism underlying endocytosis of Notch ligands have helped to identify the main players important and necessary for this process. Here, we review the Notch ligands, and the proposed models of Notch activation by Notch ligand endocytosis, highlighting key molecules involved. In particular, we discuss recent studies on Notch ligands involved in T cell development, current studies aimed at elucidating the relevance of Notch ligand endocytosis during T cell development and the identification of key players necessary for ligand endocytosis in the thymus and during T cell development.
Collapse
Affiliation(s)
- Divya K Shah
- Department of Immunology, Sunnybrook Research Institute, University of Toronto, 2075 Bayview Avenue, Toronto, ON, M4 N 3M5, Canada.
| | | |
Collapse
|
41
|
Thompson PK, Zúñiga-Pflücker JC. On becoming a T cell, a convergence of factors kick it up a Notch along the way. Semin Immunol 2011; 23:350-9. [PMID: 21981947 DOI: 10.1016/j.smim.2011.08.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 08/19/2011] [Indexed: 12/18/2022]
Abstract
The thymus is seeded by bone marrow-derived progenitors, which undergo a series of differentiation and proliferation events in order to generate functional T lymphocytes. The Notch signaling pathway, together with multiple transcription factors, act in concert to commit progenitors to a T-lineage fate, extinguishing non-T cell potential, inducing thymocyte differentiation and supporting proliferation and survival along the way to becoming a mature T cell. This review focuses on recent evidence regarding the complex interplay between the Notch pathway and other key transcription factors at specific lineage-decision points during the program of T cell development.
Collapse
Affiliation(s)
- Patrycja K Thompson
- Department of Immunology, University of Toronto, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | | |
Collapse
|
42
|
Sun JC, Lanier LL. NK cell development, homeostasis and function: parallels with CD8⁺ T cells. Nat Rev Immunol 2011; 11:645-57. [PMID: 21869816 DOI: 10.1038/nri3044] [Citation(s) in RCA: 495] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Natural killer (NK) cells survey host tissues for signs of infection, transformation or stress and, true to their name, kill target cells that have become useless or are detrimental to the host. For decades, NK cells have been classified as a component of the innate immune system. However, accumulating evidence in mice and humans suggests that, like the B and T cells of the adaptive immune system, NK cells are educated during development, possess antigen-specific receptors, undergo clonal expansion during infection and generate long-lived memory cells. In this Review, we highlight the many stages that an NK cell progresses through during its remarkable lifetime, discussing similarities and differences with its close relative, the cytotoxic CD8(+) T cell.
Collapse
Affiliation(s)
- Joseph C Sun
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | | |
Collapse
|
43
|
Huang Y, Lin L, Shanker A, Malhotra A, Yang L, Dikov MM, Carbone DP. Resuscitating cancer immunosurveillance: selective stimulation of DLL1-Notch signaling in T cells rescues T-cell function and inhibits tumor growth. Cancer Res 2011; 71:6122-31. [PMID: 21825014 DOI: 10.1158/0008-5472.can-10-4366] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Deficiencies in immune function that accumulate during cancer immunoediting lead to a progressive escape from host immunosurveillance. Therapies that correct or overcome these defects could have a powerful impact on cancer management, but current knowledge of the types and mechanisms of immune escape is still incomplete. Here, we report a novel mechanism of escape from T-cell immunity that is caused by reduction in levels of the Delta family Notch ligands DLL1 and DLL4 in hematopoietic microenvironments. An important mediator of this effect was an elevation in the levels of circulating VEGF. Selective activation of the DLL1-Notch signaling pathway in bone marrow precursors enhanced T-cell activation and inhibited tumor growth. Conversely, tumor growth led to inhibition of Delta family ligand signaling through Notch in the hematopoietic environment, resulting in suppressed T-cell function. Overall, our findings uncover a novel mechanism of tumoral immune escape and suggest that a soluble multivalent form of DLL1 may offer a generalized therapeutic intervention to stimulate T-cell immunity and suppress tumor growth.
Collapse
Affiliation(s)
- Yuhui Huang
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Jin G, Zhang F, Chan KM, Xavier Wong HL, Liu B, Cheah KSE, Liu X, Mauch C, Liu D, Zhou Z. MT1-MMP cleaves Dll1 to negatively regulate Notch signalling to maintain normal B-cell development. EMBO J 2011; 30:2281-93. [PMID: 21572390 DOI: 10.1038/emboj.2011.136] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Accepted: 04/04/2011] [Indexed: 11/09/2022] Open
Abstract
Notch signalling controls the differentiation of haematopoietic progenitor cells (HPCs). Here, we show that loss of membrane-type 1 matrix metalloproteinase (MT1-MMP, MMP14), a cell surface protease expressed in bone marrow stromal cells (BMSCs), increases Notch signalling in HPCs and specifically impairs B-lymphocyte development. When co-cultured with BMSCs in vitro, HPCs differentiation towards B lymphocytes is significantly compromised on MT1-MMP-deficient BMSCs and this defect could be completely rescued by DAPT, a specific Notch signalling inhibitor. The defective B-lymphocyte development could also be largely rescued by DAPT in vivo. MT1-MMP interacts with Notch ligand Delta-like 1 (Dll1) and promotes its cleavage on cell surface in BMSCs. Ectopic MT1-MMP cleaves Dll1 and results in diminished Notch signalling in co-cultured cells. In addition, recombinant MT1-MMP cleaves a synthetic Dll1 peptide at the same site where MT1-MMP cleaves Dll1 on the cell surface. Our data suggest that MT1-MMP directly cleaves Dll1 on BMSCs to negatively regulate Notch signalling to specifically maintain normal B-cell development in bone marrow.
Collapse
Affiliation(s)
- Guoxiang Jin
- Department of Biochemistry, Center for Reproduction, Development and Growth, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Varnum-Finney B, Halasz LM, Sun M, Gridley T, Radtke F, Bernstein ID. Notch2 governs the rate of generation of mouse long- and short-term repopulating stem cells. J Clin Invest 2011; 121:1207-16. [PMID: 21285514 DOI: 10.1172/jci43868] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 12/08/2010] [Indexed: 01/17/2023] Open
Abstract
HSCs either self-renew or differentiate to give rise to multipotent cells whose progeny provide blood cell precursors. However, surprisingly little is known about the factors that regulate this choice of self-renewal versus differentiation. One candidate is the Notch signaling pathway, with ex vivo studies suggesting that Notch regulates HSC differentiation, although a functional role for Notch in HSC self-renewal in vivo remains controversial. Here, we have shown that Notch2, and not Notch1, inhibits myeloid differentiation and enhances generation of primitive Sca-1(+)c-kit(+) progenitors following in vitro culture of enriched HSCs with purified Notch ligands. In mice, Notch2 enhanced the rate of formation of short-term repopulating multipotential progenitor cells (MPPs) as well as long-term repopulating HSCs, while delaying myeloid differentiation in BM following injury. However, consistent with previous reports, once homeostasis was achieved, neither Notch1 nor Notch2 affected repopulating cell self-renewal. These data indicate a Notch2-dependent role in assuring orderly repopulation by HSCs, MPPs, myeloid cells, and lymphoid cells during BM regeneration.
Collapse
Affiliation(s)
- Barbara Varnum-Finney
- Pediatric Oncology, Clinical Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
Notch signaling critically mediates various hematopoietic lineage decisions and is induced in mammals by Notch ligands that are classified into 2 families, Delta-like (Delta-like-1, -3 and -4) and Jagged (Jagged1 and Jagged2), based on structural homology with both Drosophila ligands Delta and Serrate, respectively. Because the functional differences between mammalian Notch ligands were still unclear, we have investigated their influence on early human hematopoiesis and show that Jagged2 affects hematopoietic lineage decisions very similarly as Delta-like-1 and -4, but very different from Jagged1. OP9 coculture experiments revealed that Jagged2, like Delta-like ligands, induces T-lineage differentiation and inhibits B-cell and myeloid development. However, dose-dependent Notch activation studies, gene expression analysis, and promoter activation assays indicated that Jagged2 is a weaker Notch1-activator compared with the Delta-like ligands, revealing a Notch1 specific signal strength hierarchy for mammalian Notch ligands. Strikingly, Lunatic-Fringe- mediated glycosylation of Notch1 potentiated Notch signaling through Delta-like ligands and also Jagged2, in contrast to Jagged1. Thus, our results reveal a unique role for Jagged1 in preventing the induction of T-lineage differentiation in hematopoietic stem cells and show an unexpected functional similarity between Jagged2 and the Delta-like ligands.
Collapse
|
47
|
Brown G, Hughes PJ, Michell RH, Ceredig R. The versatility of haematopoietic stem cells: implications for leukaemia. Crit Rev Clin Lab Sci 2010; 47:171-80. [DOI: 10.3109/10408363.2010.530150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Geoffrey Brown
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| | | | | | | |
Collapse
|
48
|
Hoyne GF, Chapman G, Sontani Y, Pursglove SE, Dunwoodie SL. A cell autonomous role for the Notch ligand Delta-like 3 in αβ T-cell development. Immunol Cell Biol 2010; 89:696-705. [PMID: 21151194 DOI: 10.1038/icb.2010.154] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Notch signalling is critical to help direct T-cell lineage commitment in early T-cell progenitors and in the development of αβ T-cells. Epithelial and stromal cell populations in the thymus express the Notch DSL (Delta, Serrate and Lag2)ligands Delta-like 1 (Dll1), Delta-like 4 (Dll4), Jagged 1 and Jagged 2, and induce Notch signalling in thymocytes that express the Notch receptor. At present there is nothing known about the role of the Delta-like 3 (Dll3) ligand in the immune system. Here we describe a novel cell autonomous role for Dll3 in αβ T-cell development. We show that Dll3 cannot activate Notch when expressed in trans but like other Notch ligands it can inhibit Notch signalling when expressed in cis with the receptor. The loss of Dll3 leads to an increase in Hes5 expression in double positive thymocytes and their increased production of mature CD4(+) and CD8(+) T cells. Studies using competitive irradiation chimeras proved that Dll3 acts in a cell autonomous manner to regulate positive selection but not negative selection of autoreactive T cells. Our results indicate that Dll3 has a unique function during T-cell development that is distinct from the role played by the other DSL ligands of Notch and is in keeping with other recent studies indicating that Dll1 and Dll3 ligands have non-overlapping roles during embryonic development.
Collapse
Affiliation(s)
- Gerard F Hoyne
- The Laboratory of T Cell Development and Regulation, John Curtin School of Medical Research, Australian National University Canberra, Canberra, Australian Capital Territory, Australia.
| | | | | | | | | |
Collapse
|
49
|
Lunatic Fringe prolongs Delta/Notch-induced self-renewal of committed αβ T-cell progenitors. Blood 2010; 117:1184-95. [PMID: 21097675 DOI: 10.1182/blood-2010-07-296616] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Lunatic Fringe (Lfng) enhances Notch1 activation by Delta-like 4 (DL4) to promote Notch1-dependent T-lineage commitment of thymus-seeding progenitors. Subsequently, Notch1 and T-cell receptor-β (TCRβ)-containing pre-TCR complexes signal CD4/CD8 double-negative 3 (DN3) committed T-cell progenitors to survive, proliferate, and differentiate into CD4/CD8 double-positive (DP) αβ T-cell precursors. Few DP thymocytes develop without Notch1 or pre-TCR signals, whereas ectopic Notch1 activation causes T-cell leukemia. However, mechanisms of a Notch-pre-TCR collaboration during this "β-selection" process are poorly understood. We genetically manipulated Lfng to attenuate or enhance Notch1 activation in DN3 thymocytes without inducing leukemogenesis. We show that Lfng temporally sustains DL-induced Notch1 signaling to prolong proliferative self-renewal of pre-DP thymocytes. Pre-TCR signaling greatly augmented Notch trophic functions to promote robust proliferation of pre-DP progenitors. In contrast, in the absence of DL/Notch signaling, pre-TCR-expressing progenitors rapidly atrophied and differentiated into DP thymocytes. Thus, Lfng prolongs Notch1 signaling to promote self-renewal more than differentiation during the early stages of β-selection. Our data provide novel insights into the Notch-pre-TCR collaboration, and suggest that decreasing Lfng expression during the DN3-DP transition minimizes the potent leukemogenic potential of Notch1 signaling.
Collapse
|
50
|
Targeting the mechanisms of resistance to chemotherapy and radiotherapy with the cancer stem cell hypothesis. JOURNAL OF ONCOLOGY 2010; 2011:941876. [PMID: 20981352 PMCID: PMC2958340 DOI: 10.1155/2011/941876] [Citation(s) in RCA: 157] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Accepted: 09/14/2010] [Indexed: 02/06/2023]
Abstract
Despite advances in treatment, cancer remains the 2nd most common cause of death in the United States. Poor cure rates may result from the ability of cancer to recur and spread after initial therapies have seemingly eliminated detectable signs of disease. A growing body of evidence supports a role for cancer stem cells (CSCs) in tumor regrowth and spread after initial treatment. Thus, targeting CSCs in combination with traditional induction therapies may improve treatment outcomes and survival rates. Unfortunately, CSCs tend to be resistant to chemo- and radiation therapy, and a better understanding of the mechanisms underlying CSC resistance to treatment is necessary. This paper provides an update on evidence that supports a fundamental role for CSCs in cancer progression, summarizes potential mechanisms of CSC resistance to treatment, and discusses classes of drugs currently in preclinical or clinical testing that show promise at targeting CSCs.
Collapse
|