1
|
Belinsky G, Ruan J, Fattahi N, Mehta S, Boddupalli CS, Mistry PK, Nair S. Modeling bone marrow microenvironment and hematopoietic dysregulation in Gaucher disease through VavCre mediated Gba deletion. Hum Mol Genet 2025; 34:952-966. [PMID: 40197748 PMCID: PMC12085781 DOI: 10.1093/hmg/ddaf045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/13/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
Biallelic mutations in Gba cause Gaucher disease (GD), a lysosomal disorder characterized by deficient glucocerebrosidase activity and the accumulation of glucosylceramide (GlcCer) and glucosylsphingosine (GlcSph), primarily in macrophages. Beyond macrophages, GD pathology affects additional hematopoietic lineages, contributing to immune dysregulation. Existing Mx1-Cre Gba knockout models require induction protocols that lead to gene deletion outside hematopoietic cells, limiting the study of hematopoietic-specific effects. To overcome these limitations, we generated a hematopoietic-specific Gba knockout model by crossing Gbafl/fl mice with Vav-Cre, enabling early deletion of Gba exons 8-11 in hematopoietic stem and progenitor cells. These mice were backcrossed to 129X1/SvJ and C57BL/6 J backgrounds, revealing that genetic background significantly influences disease severity. Efficient Gba excision was confirmed in bone marrow, spleen, and thymus, with minimal recombination in the liver. In VavCre 129 GD mice, glucocerebrosidase activity in the spleen was severely reduced, leading to GlcCer and GlcSph accumulation and Gaucher cell infiltration in the spleen and femurs. Transcriptomic analysis identified upregulation of inflammatory and lysosomal pathways. Immune cell deconvolution from RNA-seq data further revealed an expansion of monocytes, dendritic cells, and pro-inflammatory macrophage subsets, suggesting an altered immune landscape. Additionally, GPNMB, a potential GD biomarker, was significantly elevated in both spleens and sera of VavCre 129 GD mice. This hematopoietic-specific GD model provides a powerful platform for studying GD pathophysiology, modifier genes, and immune dysregulation. It offers new opportunities for biomarker discovery and for developing strategies targeting hematopoietic and immune mechanisms in GD and related lysosomal storage disorders.
Collapse
Affiliation(s)
- Glenn Belinsky
- Department of Medicine (Digestive Diseases), Yale School of Medicine, The Anylan Center, 300 Cedar St, New Haven, Connecticut 06519, United States
| | - Jiapeng Ruan
- Department of Medicine (Digestive Diseases), Yale School of Medicine, The Anylan Center, 300 Cedar St, New Haven, Connecticut 06519, United States
| | - Nima Fattahi
- Department of Medicine (Digestive Diseases), Yale School of Medicine, The Anylan Center, 300 Cedar St, New Haven, Connecticut 06519, United States
| | - Sameet Mehta
- Yale Center for Genome Analysis, Department of Medicine (Digestive Diseases), Yale School of Medicine, The Anylan Center, 300 Cedar St, New Haven, Connecticut 06519, United States
| | - Chandra Sekhar Boddupalli
- Department of Medicine (Digestive Diseases), Yale School of Medicine, The Anylan Center, 300 Cedar St, New Haven, Connecticut 06519, United States
| | - Pramod K Mistry
- Department of Medicine (Digestive Diseases), Yale School of Medicine, The Anylan Center, 300 Cedar St, New Haven, Connecticut 06519, United States
| | - Shiny Nair
- Department of Medicine (Digestive Diseases), Yale School of Medicine, The Anylan Center, 300 Cedar St, New Haven, Connecticut 06519, United States
| |
Collapse
|
2
|
Guo Y, Zhu M, Yu Z, Li Q, Chen Y, Ci L, Sun R, Shen R. Generation and characterization of a tamoxifen-inducible lineage tracing tool Cd2-P2A-CreERT2 knock-in mice. Front Immunol 2025; 16:1482070. [PMID: 40129982 PMCID: PMC11931051 DOI: 10.3389/fimmu.2025.1482070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
Introduction The new targeted gene editing technologies, such as the CRISPR/Cas system, enable researchers to insert or delete genes at targeted loci efficiently. The Cre-loxp recombination system is widely used to activate or inactivate genes with high spatial and temporal specificity. Methods Using the CRISPR/Cas9 system, we inserted the CreERT2 transgene expression cassette into the Cd2 gene locus to generate conditional Cre-driver line Cd2-CreERT2 knock-in mice, which drove the expression of CreERT2 by the endogenous Cd2 promoter. By mating the Cd2-CreERT2 strain with a Rosa26-LSL-tdTomato reporter mouse strain which contains a tdTomato expression fragment blocked with a loxP-flanked STOP cassette (LSL) driven by a CAG promoter, a Cd2-CreERT2;Rosa26-LSL-tdTomato reporter strain was obtained to evaluate the expression pattern of CD2 in different cell types. Results After treatment with tamoxifen, the Cd2-CreERT2 knock-in mice were induced to perform efficient recombination at the loxP site following CreERT2 activation and cause the expression of tdTomato fluorescence. The tdTomato and CD2 were expressed in the T cells of peripheral blood, spleen and mesenteric lymph nodes, whereas detected in a low proportion in the B cells. While about 20% of cells labeled with tamoxifen-induced tdTomato were CD2+ monocytes in peripheral blood, 10% of dendritic cells were tdTomato+/CD2+ cells. Tamoxifen-independent expression of tdTomato occurred in approximately 3% of CD2+ macrophages, but in negligible (~0.5%) in CD2+ granulocytes. Discussion This work supplied a new transgenic mouse as a valuable tool for lineage tracing in CD2-expressing cells, for conditional mutant studies of immune modulatory effects in a time-dependent manner, and analysis of the potential therapeutic effect of CD2-targeting biologics.
Collapse
Affiliation(s)
- Yang Guo
- Model Organism R&D Department, Shanghai Laboratory Animal Research Center, Shanghai, China
| | - Mengyan Zhu
- Model Organism R&D Department, Shanghai Laboratory Animal Research Center, Shanghai, China
| | - Zhilan Yu
- Model Organism R&D Department, Shanghai Laboratory Animal Research Center, Shanghai, China
| | - Qing Li
- Shanghai Engineering Research Center for Model Organizations, Shanghai Model Organisms Center, Inc., Shanghai, China
| | - Yanjuan Chen
- Model Organism R&D Department, Shanghai Laboratory Animal Research Center, Shanghai, China
| | - Lei Ci
- Shanghai Engineering Research Center for Model Organizations, Shanghai Model Organisms Center, Inc., Shanghai, China
| | - Ruilin Sun
- Shanghai Engineering Research Center for Model Organizations, Shanghai Model Organisms Center, Inc., Shanghai, China
| | - Ruling Shen
- Model Organism R&D Department, Shanghai Laboratory Animal Research Center, Shanghai, China
| |
Collapse
|
3
|
Li M, Zhou X, Li Y, Zhu X, Li Y, Hitosugi T, Zeng H. CPT2-mediated Fatty Acid Oxidation Is Dispensable for Humoral Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1139-1149. [PMID: 39258879 PMCID: PMC11458349 DOI: 10.4049/jimmunol.2400285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/16/2024] [Indexed: 09/12/2024]
Abstract
B cell activation is accompanied by dynamic metabolic reprogramming, supported by a multitude of nutrients that include glucose, amino acids, and fatty acids. Although several studies have indicated that fatty acid mitochondrial oxidation is critical for immune cell functions, contradictory findings have been reported. Carnitine palmitoyltransferase II (CPT2) is a critical enzyme for long-chain fatty acid oxidation in mitochondria. In this study, we test the requirement of CPT2 for humoral immunity using a mouse model with a lymphocyte-specific deletion of CPT2. Stable [13C] isotope tracing reveals highly reduced fatty acid-derived citrate production in CPT2-deficient B cells. Yet, CPT2 deficiency has no significant impact on B cell development, B cell activation, germinal center formation, and Ab production upon either thymus-dependent or -independent Ag challenges. Together, our findings indicate that CPT2-mediated fatty acid oxidation is dispensable for humoral immunity, highlighting the metabolic flexibility of lymphocytes.
Collapse
Affiliation(s)
- Meilu Li
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, P. R. China
| | - Xian Zhou
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
| | - Yanfeng Li
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
| | - Xingxing Zhu
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
| | - Yuzhen Li
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, P. R. China
| | - Taro Hitosugi
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Hu Zeng
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic Rochester, MN 55905, USA
| |
Collapse
|
4
|
Li M, Zhou X, Zhu X, Li Y, Hitosugi T, Li Y, Zeng H. CPT2 mediated fatty acid oxidation is dispensable for humoral immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594133. [PMID: 38798358 PMCID: PMC11118297 DOI: 10.1101/2024.05.15.594133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
B cell activation is accompanied by dynamic metabolic reprogramming, supported by a multitude of nutrients that include glucose, amino acids and fatty acids. While several studies have indicated that fatty acid mitochondrial oxidation is critical for immune cell functions, contradictory findings have been reported. Carnitine palmitoyltransferase II (CPT2) is a critical enzyme for long-chain fatty acid oxidation in mitochondria. Here, we test the requirement of CPT2 for humoral immunity using a mouse model with a lymphocyte specific deletion of CPT2. Stable 13C isotope tracing reveals highly reduced fatty acid-derived citrate production in CPT2 deficient B cells. Yet, CPT2 deficiency has no significant impact on B cell development, B cell activation, germinal center formation, and antibody production upon either thymus-dependent or -independent antigen challenges. Together, our findings indicate that CPT2 mediated fatty acid oxidation is dispensable for humoral immunity, highlighting the metabolic flexibility of lymphocytes.
Collapse
Affiliation(s)
- Meilu Li
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, P. R. China
| | - Xian Zhou
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
| | - Xingxing Zhu
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
| | - Yanfeng Li
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
| | - Taro Hitosugi
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Yuzhen Li
- Department of Dermatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, P. R. China
| | - Hu Zeng
- Division of Rheumatology, Department of Medicine, Mayo Clinic Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic Rochester, MN 55905, USA
| |
Collapse
|
5
|
Crespiatico I, Zaghi M, Mastini C, D'Aliberti D, Mauri M, Mercado CM, Fontana D, Spinelli S, Crippa V, Inzoli E, Manghisi B, Civettini I, Ramazzotti D, Sangiorgio V, Gengotti M, Brambilla V, Aroldi A, Banfi F, Barone C, Orsenigo R, Riera L, Riminucci M, Corsi A, Breccia M, Morotti A, Cilloni D, Roccaro A, Sacco A, Stagno F, Serafini M, Mottadelli F, Cazzaniga G, Pagni F, Chiarle R, Azzoni E, Sessa A, Gambacorti-Passerini C, Elli EM, Mologni L, Piazza R. First-hit SETBP1 mutations cause a myeloproliferative disorder with bone marrow fibrosis. Blood 2024; 143:1399-1413. [PMID: 38194688 DOI: 10.1182/blood.2023021349] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/11/2024] Open
Abstract
ABSTRACT SETBP1 mutations are found in various clonal myeloid disorders. However, it is unclear whether they can initiate leukemia, because SETBP1 mutations typically appear as later events during oncogenesis. To answer this question, we generated a mouse model expressing mutated SETBP1 in hematopoietic tissue: this model showed profound alterations in the differentiation program of hematopoietic progenitors and developed a myeloid neoplasm with megakaryocytic dysplasia, splenomegaly, and bone marrow fibrosis, prompting us to investigate SETBP1 mutations in a cohort of 36 triple-negative primary myelofibrosis (TN-PMF) cases. We identified 2 distinct subgroups, one carrying SETBP1 mutations and the other completely devoid of somatic variants. Clinically, a striking difference in disease aggressiveness was noted, with patients with SETBP1 mutation showing a much worse clinical course. In contrast to myelodysplastic/myeloproliferative neoplasms, in which SETBP1 mutations are mostly found as a late clonal event, single-cell clonal hierarchy reconstruction in 3 patients with TN-PMF from our cohort revealed SETBP1 to be a very early event, suggesting that the phenotype of the different SETBP1+ disorders may be shaped by the opposite hierarchy of the same clonal SETBP1 variants.
Collapse
Affiliation(s)
- Ilaria Crespiatico
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Mattia Zaghi
- Neuroepigenetics Unit, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Mastini
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Deborah D'Aliberti
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Mario Mauri
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Carl Mirko Mercado
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Diletta Fontana
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Silvia Spinelli
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Valentina Crippa
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Elena Inzoli
- Hematology Division and Bone Marrow Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico San Gerardo dei Tintori, Monza, Italy
| | - Beatrice Manghisi
- Hematology Division and Bone Marrow Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico San Gerardo dei Tintori, Monza, Italy
| | - Ivan Civettini
- Hematology Division and Bone Marrow Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico San Gerardo dei Tintori, Monza, Italy
| | - Daniele Ramazzotti
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Valentina Sangiorgio
- Hematology Division and Bone Marrow Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico San Gerardo dei Tintori, Monza, Italy
| | - Michele Gengotti
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | | | - Andrea Aroldi
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Hematology Division and Bone Marrow Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico San Gerardo dei Tintori, Monza, Italy
| | - Federica Banfi
- Neuroepigenetics Unit, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Cristiana Barone
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Roberto Orsenigo
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory, Vall d'Hebron Research Institute, Vall d'Hebron Hospital Barcelona UAB, Barcelona, Spain
| | - Ludovica Riera
- Department of Pathology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Azienda Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Alessandro Corsi
- Department of Molecular Medicine, Azienda Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Massimo Breccia
- Department of Translational and Precision Medicine, Azienda Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Turin, Italy
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Turin, Italy
| | - Aldo Roccaro
- Clinical Trial Center, Translational Research and Phase I Unit, Azienda Socio Sanitaria Territoriale Spedali Civili di Brescia, Brescia, Italy
| | - Antonio Sacco
- Clinical Trial Center, Translational Research and Phase I Unit, Azienda Socio Sanitaria Territoriale Spedali Civili di Brescia, Brescia, Italy
| | - Fabio Stagno
- Division of Hematology, Azienda Ospedaliero Universitaria Policlinico G. Rodolico-S. Marco, Catania, Italy
| | - Marta Serafini
- Centro Tettamanti, Istituto di Ricovero e Cura a Carattere Scientifico San Gerardo dei Tintori, Monza, Italy
| | - Federica Mottadelli
- Centro Tettamanti, Istituto di Ricovero e Cura a Carattere Scientifico San Gerardo dei Tintori, Monza, Italy
| | - Giovanni Cazzaniga
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Centro Tettamanti, Istituto di Ricovero e Cura a Carattere Scientifico San Gerardo dei Tintori, Monza, Italy
| | - Fabio Pagni
- Department of Pathology, University of Milan-Bicocca, Monza, Italy
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Italy
- Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA
- European Institute of Oncology Istituto di Ricovero e Cura a Carattere Scientifico, Division of Haematopathology, Milan, Italy
| | - Emanuele Azzoni
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Alessandro Sessa
- Neuroepigenetics Unit, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Carlo Gambacorti-Passerini
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Hematology Division and Bone Marrow Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico San Gerardo dei Tintori, Monza, Italy
| | - Elena Maria Elli
- Hematology Division and Bone Marrow Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico San Gerardo dei Tintori, Monza, Italy
| | - Luca Mologni
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Hematology Division and Bone Marrow Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
6
|
Chen J, Wen Y, Lin L, Cui Y, Chen Z, Gao J, Zhuang Y, Chen Q. Fosl2 Deficiency Predisposes Mice to Osteopetrosis, Leading to Bone Marrow Failure. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1081-1093. [PMID: 38380993 DOI: 10.4049/jimmunol.2300592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/16/2024] [Indexed: 02/22/2024]
Abstract
Arthritis causes Fos-like 2 (Fosl2) inactivation, and various immune cells contribute to its pathogenesis. However, little is known about the role of Fosl2 in hematopoiesis and the possible pathological role of Fosl2 inactivation in the hematopoietic system in arthritis. In this study, we show that Fosl2 maintains hematopoietic stem cell (HSC) quiescence and differentiation while controlling the inflammatory response via macrophages. Fosl2-specific deletion in the hematopoietic system caused the expansion of HSCs and myeloid cell growth while affecting erythroid and B cell differentiation. Fosl2 inactivation enhanced macrophage M1 polarization and stimulated proinflammatory cytokines and myeloid growth factors, skewing HSCs toward myeloid cell differentiation, similar to hematopoietic alterations in arthritic mice. Loss of Fosl2 mediated by Vav-iCre also displays an unexpected deletion in embryonic erythro-myeloid progenitor-derived osteoclasts, leading to osteopetrosis and anemia. The reduced bone marrow cellularity in Vav-iCreFosl2f/f mice is a consequence of the reduced bone marrow space in osteopetrotic mice rather than a direct role of Fosl2 in hematopoiesis. Thus, Fosl2 is indispensable for erythro-myeloid progenitor-derived osteoclasts to maintain the medullary cavity to ensure normal hematopoiesis. These findings improve our understanding of the pathogenesis of bone-destructive diseases and provide important implications for developing therapeutic approaches for these diseases.
Collapse
Affiliation(s)
- Jinfeng Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China
| | - Yi Wen
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Lili Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China
| | - Yuchen Cui
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China
| | - Zhenyu Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China
| | - Jing Gao
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China
| | - Yifang Zhuang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, Fujian, China
| |
Collapse
|
7
|
Limone F, Couto A, Wang JY, Zhang Y, McCourt B, Huang C, Minkin A, Jani M, McNeer S, Keaney J, Gillet G, Gonzalez RL, Goodman WA, Kadiu I, Eggan K, Burberry A. Myeloid and lymphoid expression of C9orf72 regulates IL-17A signaling in mice. Sci Transl Med 2024; 16:eadg7895. [PMID: 38295187 PMCID: PMC11247723 DOI: 10.1126/scitranslmed.adg7895] [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: 01/26/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
A mutation in C9ORF72 is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Patients with ALS or FTD often develop autoimmunity and inflammation that precedes or coincides with the onset of neurological symptoms, but the underlying mechanisms are poorly understood. Here, we knocked out murine C9orf72 in seven hematopoietic progenitor compartments by conditional mutagenesis and found that myeloid lineage C9orf72 prevents splenomegaly, loss of tolerance, and premature mortality. Furthermore, we demonstrated that C9orf72 plays a role in lymphoid cells to prevent interleukin-17A (IL-17A) production and neutrophilia. Mass cytometry identified early and sustained elevation of the costimulatory molecule CD80 expressed on C9orf72-deficient mouse macrophages, monocytes, and microglia. Enrichment of CD80 was similarly observed in human spinal cord microglia from patients with C9ORF72-mediated ALS compared with non-ALS controls. Single-cell RNA sequencing of murine spinal cord, brain cortex, and spleen demonstrated coordinated induction of gene modules related to antigen processing and presentation and antiviral immunity in C9orf72-deficient endothelial cells, microglia, and macrophages. Mechanistically, C9ORF72 repressed the trafficking of CD80 to the cell surface in response to Toll-like receptor agonists, interferon-γ, and IL-17A. Deletion of Il17a in C9orf72-deficient mice prevented CD80 enrichment in the spinal cord, reduced neutrophilia, and reduced gut T helper type 17 cells. Last, systemic delivery of an IL-17A neutralizing antibody augmented motor performance and suppressed neuroinflammation in C9orf72-deficient mice. Altogether, we show that C9orf72 orchestrates myeloid costimulatory potency and provide support for IL-17A as a therapeutic target for neuroinflammation associated with ALS or FTD.
Collapse
Affiliation(s)
- Francesco Limone
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
- Leiden University Medical Center, LUMC, 2333 ZA Leiden, The Netherlands
| | - Alexander Couto
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Jin-Yuan Wang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Yingying Zhang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Blake McCourt
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Cerianne Huang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Adina Minkin
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Marghi Jani
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Sarah McNeer
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - James Keaney
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Gaëlle Gillet
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Rodrigo Lopez Gonzalez
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44196, USA
| | - Wendy A. Goodman
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Irena Kadiu
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Aaron Burberry
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| |
Collapse
|
8
|
Cao H, Naik SH, Amann-Zalcenstein D, Hickey P, Salim A, Cao B, Nilsson SK, Keightley MC, Lieschke GJ. Late fetal hematopoietic failure results from ZBTB11 deficiency despite abundant HSC specification. Blood Adv 2023; 7:6506-6519. [PMID: 37567157 PMCID: PMC10632610 DOI: 10.1182/bloodadvances.2022009580] [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: 12/19/2022] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/13/2023] Open
Abstract
Hematopoiesis produces diverse blood cell lineages to meet the basal needs and sudden demands of injury or infection. A rapid response to such challenges requires the expansion of specific lineages and a prompt return to balanced steady-state levels, necessitating tightly coordinated regulation. Previously we identified a requirement for the zinc finger and broad complex, tramtrak, bric-a-brac domain-containing 11 (ZBTB11) transcription factor in definitive hematopoiesis using a forward genetic screen for zebrafish myeloid mutants. To understand its relevance to mammalian systems, we extended these studies to mice. When Zbtb11 was deleted in the hematopoietic compartment, embryos died at embryonic day (E) 18.5 with hematopoietic failure. Zbtb11 hematopoietic knockout (Zbtb11hKO) hematopoietic stem cells (HSCs) were overabundantly specified from E14.5 to E17.5 compared with those in controls. Overspecification was accompanied by loss of stemness, inability to differentiate into committed progenitors and mature lineages in the fetal liver, failure to seed fetal bone marrow, and total hematopoietic failure. The Zbtb11hKO HSCs did not proliferate in vitro and were constrained in cell cycle progression, demonstrating the cell-intrinsic role of Zbtb11 in proliferation and cell cycle regulation in mammalian HSCs. Single-cell RNA sequencing analysis identified that Zbtb11-deficient HSCs were underrepresented in an erythroid-primed subpopulation and showed downregulation of oxidative phosphorylation pathways and dysregulation of genes associated with the hematopoietic niche. We identified a cell-intrinsic requirement for Zbtb11-mediated gene regulatory networks in sustaining a pool of maturation-capable HSCs and progenitor cells.
Collapse
Affiliation(s)
- Huimin Cao
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia
| | - Shalin H. Naik
- Department of Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Single Cell Open Research Endeavour, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Daniela Amann-Zalcenstein
- Single Cell Open Research Endeavour, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Advanced Genomics Facility, Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Peter Hickey
- Single Cell Open Research Endeavour, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Advanced Genomics Facility, Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Agus Salim
- Mathematics and Statistics, La Trobe University, Bundoora, VIC, Australia
- Melbourne School of Population and Global Health, School of Mathematics and Statistics, University of Melbourne, Parkville, VIC, Australia
| | - Benjamin Cao
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia
| | - Susan K. Nilsson
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, VIC, Australia
| | - M. Cristina Keightley
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
- Rural Clinical Sciences, La Trobe Rural Health School, Bendigo, VIC, Australia
| | - Graham J. Lieschke
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
9
|
Sulczewski FB, Maqueda-Alfaro RA, Alcántara-Hernández M, Perez OA, Saravanan S, Yun TJ, Seong D, Arroyo Hornero R, Raquer-McKay HM, Esteva E, Lanzar ZR, Leylek RA, Adams NM, Das A, Rahman AH, Gottfried-Blackmore A, Reizis B, Idoyaga J. Transitional dendritic cells are distinct from conventional DC2 precursors and mediate proinflammatory antiviral responses. Nat Immunol 2023; 24:1265-1280. [PMID: 37414907 PMCID: PMC10683792 DOI: 10.1038/s41590-023-01545-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 05/26/2023] [Indexed: 07/08/2023]
Abstract
High-dimensional approaches have revealed heterogeneity amongst dendritic cells (DCs), including a population of transitional DCs (tDCs) in mice and humans. However, the origin and relationship of tDCs to other DC subsets has been unclear. Here we show that tDCs are distinct from other well-characterized DCs and conventional DC precursors (pre-cDCs). We demonstrate that tDCs originate from bone marrow progenitors shared with plasmacytoid DCs (pDCs). In the periphery, tDCs contribute to the pool of ESAM+ type 2 DCs (DC2s), and these DC2s have pDC-related developmental features. Different from pre-cDCs, tDCs have less turnover, capture antigen, respond to stimuli and activate antigen-specific naïve T cells, all characteristics of differentiated DCs. Different from pDCs, viral sensing by tDCs results in IL-1β secretion and fatal immune pathology in a murine coronavirus model. Our findings suggest that tDCs are a distinct pDC-related subset with a DC2 differentiation potential and unique proinflammatory function during viral infections.
Collapse
Affiliation(s)
- Fernando Bandeira Sulczewski
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Raul A Maqueda-Alfaro
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Marcela Alcántara-Hernández
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Oriana A Perez
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Sanjana Saravanan
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Tae Jin Yun
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - David Seong
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Rebeca Arroyo Hornero
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Hayley M Raquer-McKay
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Eduardo Esteva
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Zachary R Lanzar
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Rebecca A Leylek
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Nicholas M Adams
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Annesa Das
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Adeeb H Rahman
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andres Gottfried-Blackmore
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Redwood City, CA, USA
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA.
| | - Juliana Idoyaga
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
10
|
Zhang S, Audiger C, Chopin M, Nutt SL. Transcriptional regulation of dendritic cell development and function. Front Immunol 2023; 14:1182553. [PMID: 37520521 PMCID: PMC10382230 DOI: 10.3389/fimmu.2023.1182553] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023] Open
Abstract
Dendritic cells (DCs) are sentinel immune cells that form a critical bridge linking the innate and adaptive immune systems. Extensive research addressing the cellular origin and heterogeneity of the DC network has revealed the essential role played by the spatiotemporal activity of key transcription factors. In response to environmental signals DC mature but it is only following the sensing of environmental signals that DC can induce an antigen specific T cell response. Thus, whilst the coordinate action of transcription factors governs DC differentiation, sensing of environmental signals by DC is instrumental in shaping their functional properties. In this review, we provide an overview that focuses on recent advances in understanding the transcriptional networks that regulate the development of the reported DC subsets, shedding light on the function of different DC subsets. Specifically, we discuss the emerging knowledge on the heterogeneity of cDC2s, the ontogeny of pDCs, and the newly described DC subset, DC3. Additionally, we examine critical transcription factors such as IRF8, PU.1, and E2-2 and their regulatory mechanisms and downstream targets. We highlight the complex interplay between these transcription factors, which shape the DC transcriptome and influence their function in response to environmental stimuli. The information presented in this review provides essential insights into the regulation of DC development and function, which might have implications for developing novel therapeutic strategies for immune-related diseases.
Collapse
Affiliation(s)
- Shengbo Zhang
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Cindy Audiger
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Michaël Chopin
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Stephen L. Nutt
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
11
|
Rodrigues PF, Kouklas A, Cvijetic G, Bouladoux N, Mitrovic M, Desai JV, Lima-Junior DS, Lionakis MS, Belkaid Y, Ivanek R, Tussiwand R. pDC-like cells are pre-DC2 and require KLF4 to control homeostatic CD4 T cells. Sci Immunol 2023; 8:eadd4132. [PMID: 36827419 PMCID: PMC10165717 DOI: 10.1126/sciimmunol.add4132] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 02/02/2023] [Indexed: 02/26/2023]
Abstract
Plasmacytoid dendritic cells (pDCs) have been shown to play an important role during immune responses, ranging from initial viral control through the production of type I interferons to antigen presentation. However, recent studies uncovered unexpected heterogeneity among pDCs. We identified a previously uncharacterized immune subset, referred to as pDC-like cells, that not only resembles pDCs but also shares conventional DC (cDC) features. We show that this subset is a circulating precursor distinct from common DC progenitors, with prominent cDC2 potential. Our findings from human CD2-iCre and CD300c-iCre lineage tracing mouse models suggest that a substantial fraction of cDC2s originates from pDC-like cells, which can therefore be referred to as pre-DC2. This precursor subset responds to homeostatic cytokines, such as macrophage colony stimulating factor, by expanding and differentiating into cDC2 that efficiently prime T helper 17 (TH17) cells. Development of pre-DC2 into CX3CR1+ ESAM- cDC2b but not CX3CR1- ESAM+ cDC2a requires the transcription factor KLF4. Last, we show that, under homeostatic conditions, this developmental pathway regulates the immune threshold at barrier sites by controlling the pool of TH17 cells within skin-draining lymph nodes.
Collapse
Affiliation(s)
| | | | - Grozdan Cvijetic
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
- National Institute of Dental and Craniofacial Research (NIDCR), NIH, Bethesda, MD 20892, USA
| | - Nicolas Bouladoux
- Metaorganism Immunity Section, Laboratory of Host Microbiome and Immunity, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), Bethesda, MD 20892, USA
| | - Mladen Mitrovic
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
- National Institute of Dental and Craniofacial Research (NIDCR), NIH, Bethesda, MD 20892, USA
| | - Jigar V Desai
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Djalma S Lima-Junior
- Metaorganism Immunity Section, Laboratory of Host Microbiome and Immunity, National Institute of Allergy and Infectious Diseases (NIAID), National Institute of Health (NIH), Bethesda, MD 20892, USA
| | - Michail S. Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Yasmine Belkaid
- National Institute of Dental and Craniofacial Research (NIDCR), NIH, Bethesda, MD 20892, USA
| | - Robert Ivanek
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics, 4058 Basel, Switzerland
| | - Roxane Tussiwand
- National Institute of Dental and Craniofacial Research (NIDCR), NIH, Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Mar KB, Van Dyke MC, Lopez AH, Eitson JL, Fan W, Hanners NW, Evers BM, Shelton JM, Schoggins JW. LY6E protects mice from pathogenic effects of murine coronavirus and SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.25.525551. [PMID: 36747632 PMCID: PMC9900800 DOI: 10.1101/2023.01.25.525551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
LY6E is an antiviral protein that inhibits coronavirus entry. Its expression in immune cells allows mice to control murine coronavirus infection. However, it is not known which immune cell subsets mediate this control or whether LY6E protects mice from SARS-CoV-2. In this study, we used tissue-specific Cre recombinase expression to ablate Ly6e in distinct immune compartments or in all epiblast-derived cells, and bone marrow chimeras to target Ly6e in a subset of radioresistant cells. Mice lacking Ly6e in Lyz2 -expressing cells and radioresistant Vav1 -expressing cells were more susceptible to lethal murine coronavirus infection. Mice lacking Ly6e globally developed clinical disease when challenged with the Gamma (P.1) variant of SARS-CoV-2. By contrast, wildtype mice and mice lacking type I and type III interferon signaling had no clinical symptoms after SARS-CoV-2 infection. Transcriptomic profiling of lungs from SARS-CoV-2-infected wildtype and Ly6e knockout mice revealed a striking reduction of secretory cell-associated genes in infected knockout mice, including Muc5b , an airway mucin-encoding gene that may protect against SARS-CoV-2-inflicted respiratory disease. Collectively, our study reveals distinct cellular compartments in which Ly6e confers cell intrinsic antiviral effects, thereby conferring resistance to disease caused by murine coronavirus and SARS-CoV-2.
Collapse
|
13
|
Krimpenfort RA, Behr FM, Nieuwland M, de Rink I, Kerkhoven R, von Lindern M, Nethe M. E-Cadherin Expression Distinguishes Mouse from Human Hematopoiesis in the Basophil and Erythroid Lineages. Biomolecules 2022; 12:1706. [PMID: 36421719 PMCID: PMC9688100 DOI: 10.3390/biom12111706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 09/11/2024] Open
Abstract
E-cadherin is a key regulator of epithelial cell-cell adhesion, the loss of which accelerates tumor growth and invasion. E-cadherin is also expressed in hematopoietic cells as well as epithelia. The function of hematopoietic E-cadherin is, however, mostly elusive. In this study, we explored the validity of mouse models to functionally investigate the role of hematopoietic E-cadherin in human hematopoiesis. We generated a hematopoietic-specific E-cadherin knockout mouse model. In mice, hematopoietic E-cadherin is predominantly expressed within the basophil lineage, the expression of which is dispensable for the generation of basophils. However, neither E-cadherin mRNA nor protein were detected in human basophils. In contrast, human hematopoietic E-cadherin marks the erythroid lineage. E-cadherin expression in hematopoiesis thereby revealed striking evolutionary differences between the basophil and erythroid cell lineage in humans and mice. This is remarkable as E-cadherin expression in epithelia is highly conserved among vertebrates including humans and mice. Our study therefore revealed that the mouse does not represent a suitable model to study the function of E-cadherin in human hematopoiesis and an alternative means to study the role of E-cadherin in human erythropoiesis needs to be developed.
Collapse
Affiliation(s)
- Rosa A. Krimpenfort
- Sanquin Research, Landsteiner Laboratory, Academic Medical Centre, Department of Hematopoiesis, University of Amsterdam, 1066 CX Amsterdam, The Netherlands
| | - Felix M. Behr
- Sanquin Research, Landsteiner Laboratory, Academic Medical Centre, Department of Hematopoiesis, University of Amsterdam, 1066 CX Amsterdam, The Netherlands
| | - Marja Nieuwland
- Genomics Core Facility, Netherlands Cancer Institute, 1006 BE Amsterdam, The Netherlands
| | - Iris de Rink
- Genomics Core Facility, Netherlands Cancer Institute, 1006 BE Amsterdam, The Netherlands
| | - Ron Kerkhoven
- Genomics Core Facility, Netherlands Cancer Institute, 1006 BE Amsterdam, The Netherlands
| | - Marieke von Lindern
- Sanquin Research, Landsteiner Laboratory, Academic Medical Centre, Department of Hematopoiesis, University of Amsterdam, 1066 CX Amsterdam, The Netherlands
| | - Micha Nethe
- Sanquin Research, Landsteiner Laboratory, Academic Medical Centre, Department of Hematopoiesis, University of Amsterdam, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
14
|
Yin N, Jin G, Ma Y, Zhao H, Zhang G, Li MO, Peng M. SZT2 maintains hematopoietic stem cell homeostasis via nutrient-mediated mTORC1 regulation. J Clin Invest 2022; 132:146272. [PMID: 36250465 PMCID: PMC9566891 DOI: 10.1172/jci146272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 08/19/2022] [Indexed: 11/17/2022] Open
Abstract
The mTORC1 pathway coordinates nutrient and growth factor signals to maintain organismal homeostasis. Whether nutrient signaling to mTORC1 regulates stem cell function remains unknown. Here, we show that SZT2 — a protein required for mTORC1 downregulation upon nutrient deprivation — is critical for hematopoietic stem cell (HSC) homeostasis. Ablation of SZT2 in HSCs decreased the reserve and impaired the repopulating capacity of HSCs. Furthermore, ablation of both SZT2 and TSC1 — 2 repressors of mTORC1 on the nutrient and growth factor arms, respectively — led to rapid HSC depletion, pancytopenia, and premature death of the mice. Mechanistically, loss of either SZT2 or TSC1 in HSCs led to only mild elevation of mTORC1 activity and reactive oxygen species (ROS) production. Loss of both SZT2 and TSC1, on the other hand, simultaneously produced a dramatic synergistic effect, with an approximately 10-fold increase of mTORC1 activity and approximately 100-fold increase of ROS production, which rapidly depleted HSCs. These data demonstrate a critical role of nutrient mTORC1 signaling in HSC homeostasis and uncover a strong synergistic effect between nutrient- and growth factor–mediated mTORC1 regulation in stem cells.
Collapse
Affiliation(s)
- Na Yin
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Gang Jin
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Yuying Ma
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Hanfei Zhao
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Guangyue Zhang
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Ming O. Li
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, New York, USA
| | - Min Peng
- Department of Basic Medical Sciences, School of Medicine, and
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| |
Collapse
|
15
|
Wang Z, Heid B, Lu R, Sachdeva M, Edwards MR, Ren J, Cecere TE, Khan D, Jeboda T, Kirsch DG, Reilly CM, Dai R, Ahmed SA. Deletion of microRNA-183-96-182 Cluster in Lymphocytes Suppresses Anti-DsDNA Autoantibody Production and IgG Deposition in the Kidneys in C57BL/6-Fas lpr/lpr Mice. Front Genet 2022; 13:840060. [PMID: 35873462 PMCID: PMC9301314 DOI: 10.3389/fgene.2022.840060] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Dysregulated miRNAs have been implicated in the pathogenesis of systemic lupus erythematosus (SLE). Our previous study reported a substantial increase in three miRNAs located at the miR-183-96-182 cluster (miR-183C) in several autoimmune lupus-prone mice, including MRL/lpr and C57BL/6-lpr (B6/lpr). This study reports that in vitro inhibition of miR-182 alone or miR-183C by specific antagomirs in activated splenocytes from autoimmune-prone MRL/lpr and control MRL mice significantly reduced lupus-related inflammatory cytokines, interferon-gamma (IFNγ), and IL-6 production. To further characterize the role of miR-182 and miR-183C cluster in vivo in lupus-like disease and lymphocyte phenotypes, we used hCD2-iCre to generate B6/lpr mice with conditional deletion of miR-182 or miR-183C in CD2+ lymphocytes (miR-182-/-B6/lpr and miR-183C-/-B6/lpr). The miR-182-/-B6/lpr and miR-183C-/-B6/lpr mice had significantly reduced deposition of IgG immunocomplexes in the kidney when compared to their respective littermate controls, although there appeared to be no remarkable changes in renal pathology. Importantly, we observed a significant reduction of serum anti-dsDNA autoantibodies in miR-183C-/-B6/lpr mice after reaching 24 weeks-of age compared to age-matched miR-183Cfl/flB6/lpr controls. In vitro activated splenocytes from miR-182-/-B6/lpr mice and miR-183C-/-B6/lpr mice showed reduced ability to produce lupus-associated IFNγ. Forkhead box O1(Foxo1), a previously validated miR-183C miRNAs target, was increased in the splenic CD4+ cells of miR-182-/-B6/lpr and miR-183C-/-B6/lpr mice. Furthermore, in vitro inhibition of Foxo1 with siRNA in splenocytes from miR-182-/-B6/lpr and miR-183C-/-B6/lpr mice significantly increased IFNγ expression following anti-CD3/CD28 stimulation, suggesting that miR-182 and miR-183C miRNAs regulate the inflammatory IFNγ in splenocytes via targeting Foxo1. The deletion of either miR-182 alone or the whole miR-183C cluster, however, had no marked effect on the composition of T and B cell subsets in the spleens of B6/lpr mice. There were similar percentages of CD4+, CD8+, CD19+, as well as Tregs, follicular helper T (TFH), germinal center B (GCB), and plasma cells in the miR-183C-/-B6/lpr and miR-182-/-B6/lpr mice and their respective littermate controls, miR-183Cfl/flB6/lpr and miR-182fl/flB6/lpr mice. Together, our data demonstrate a role of miR-183C in the regulation of anti-dsDNA autoantibody production in vivo in B6/lpr mice and the induction of IFNγ in in vitro activated splenocytes from B6/lpr mice.
Collapse
Affiliation(s)
- Zhuang Wang
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - Bettina Heid
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - Ran Lu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - Mohit Sachdeva
- Preclinical Lead Immunology, Spark Theraprutics, Philadelphia, PA, United States
| | - Michael R. Edwards
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - JingJing Ren
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - Thomas E. Cecere
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - Deena Khan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - Taschua Jeboda
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - David G. Kirsch
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
| | - Christopher M. Reilly
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
- Edward Via College of Osteopathic Medicine, Blacksburg, VA, United States
| | - Rujuan Dai
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| | - S. Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine (VMCVM), Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
16
|
Lombard‐Vadnais F, Lacombe J, Chabot‐Roy G, Ferron M, Lesage S. OCA‐B does not act as a transcriptional coactivator in T cells. Immunol Cell Biol 2022; 100:338-351. [DOI: 10.1111/imcb.12543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 02/14/2022] [Accepted: 03/09/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Félix Lombard‐Vadnais
- Immunologie‐oncologie Centre de recherche de l’Hôpital Maisonneuve‐Rosemont Montréal QC H1T 2M4 Canada
- Department of Microbiology & Immunology McGill University Montreal QC H3A 0G4 Canada
| | - Julie Lacombe
- Molecular Physiology Research Unit Institut de recherches cliniques de Montréal Montréal QC H2W 1R7 Canada
| | - Geneviève Chabot‐Roy
- Immunologie‐oncologie Centre de recherche de l’Hôpital Maisonneuve‐Rosemont Montréal QC H1T 2M4 Canada
| | - Mathieu Ferron
- Molecular Physiology Research Unit Institut de recherches cliniques de Montréal Montréal QC H2W 1R7 Canada
- Département de médecine Université de Montréal Montréal QC H3T 1J4 Canada
- Division of Experimental Medicine McGill University Montreal QC H3A 0G4 Canada
| | - Sylvie Lesage
- Immunologie‐oncologie Centre de recherche de l’Hôpital Maisonneuve‐Rosemont Montréal QC H1T 2M4 Canada
- Département de microbiologie, infectiologie et immunologie Université de Montréal Montréal QC H3T 1J4 Canada
| |
Collapse
|
17
|
Feng J, Pucella JN, Jang G, Alcántara-Hernández M, Upadhaya S, Adams NM, Khodadadi-Jamayran A, Lau CM, Stoeckius M, Hao S, Smibert P, Tsirigos A, Idoyaga J, Reizis B. Clonal lineage tracing reveals shared origin of conventional and plasmacytoid dendritic cells. Immunity 2022; 55:405-422.e11. [PMID: 35180378 PMCID: PMC9344860 DOI: 10.1016/j.immuni.2022.01.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 09/23/2021] [Accepted: 01/24/2022] [Indexed: 12/14/2022]
Abstract
Developmental origins of dendritic cells (DCs) including conventional DCs (cDCs, comprising cDC1 and cDC2 subsets) and plasmacytoid DCs (pDCs) remain unclear. We studied DC development in unmanipulated adult mice using inducible lineage tracing combined with clonal DNA "barcoding" and single-cell transcriptome and phenotype analysis (CITE-seq). Inducible tracing of Cx3cr1+ hematopoietic progenitors in the bone marrow showed that they simultaneously produce all DC subsets including pDCs, cDC1s, and cDC2s. Clonal tracing of hematopoietic stem cells (HSCs) and of Cx3cr1+ progenitors revealed clone sharing between cDC1s and pDCs, but not between the two cDC subsets or between pDCs and B cells. Accordingly, CITE-seq analyses of differentiating HSCs and Cx3cr1+ progenitors identified progressive stages of pDC development including Cx3cr1+ Ly-6D+ pro-pDCs that were distinct from lymphoid progenitors. These results reveal the shared origin of pDCs and cDCs and suggest a revised scheme of DC development whereby pDCs share clonal relationship with cDC1s.
Collapse
Affiliation(s)
- Jue Feng
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA.
| | - Joseph N Pucella
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Geunhyo Jang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Marcela Alcántara-Hernández
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Immunology Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Samik Upadhaya
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Nicholas M Adams
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Alireza Khodadadi-Jamayran
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Colleen M Lau
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Marlon Stoeckius
- Technology Innovation Laboratory, New York Genome Center, New York, NY 10013, USA
| | - Stephanie Hao
- Technology Innovation Laboratory, New York Genome Center, New York, NY 10013, USA
| | - Peter Smibert
- Technology Innovation Laboratory, New York Genome Center, New York, NY 10013, USA
| | - Aristotelis Tsirigos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Juliana Idoyaga
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Immunology Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
18
|
Hagman JR, Arends T, Laborda C, Knapp JR, Harmacek L, O'Connor BP. Chromodomain helicase DNA-binding 4 (CHD4) regulates early B cell identity and V(D)J recombination. Immunol Rev 2021; 305:29-42. [PMID: 34927255 DOI: 10.1111/imr.13054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/22/2021] [Accepted: 12/02/2021] [Indexed: 12/20/2022]
Abstract
B lymphocytes develop from uncommitted precursors into immunoglobulin (antibody)-producing B cells, a major arm of adaptive immunity. Progression of early progenitors to antibody-expressing cells in the bone marrow is orchestrated by the temporal regulation of different gene programs at discrete developmental stages. A major question concerns how B cells control the accessibility of these genes to transcription factors. Research has implicated nucleosome remodeling ATPases as mediators of chromatin accessibility. Here, we describe studies of chromodomain helicase DNA-binding 4 (CHD4; also known as Mi-2β) in early B cell development. CHD4 comprises multiple domains that function in nucleosome mobilization and histone binding. CHD4 is a key component of Nucleosome Remodeling and Deacetylase, or NuRD (Mi-2) complexes, which assemble with other proteins that mediate transcriptional repression. We review data demonstrating that CHD4 is necessary for B lineage identity: early B lineage progression, proliferation in response to interleukin-7, responses to DNA damage, and cell survival in vivo. CHD4-NuRD is also required for the Ig heavy-chain repertoire by promoting utilization of distal variable (VH ) gene segments in V(D)J recombination. In conclusion, the regulation of chromatin accessibility by CHD4 is essential for production of antibodies by B cells, which in turn mediate humoral immune responses to pathogens and disease.
Collapse
Affiliation(s)
- James R Hagman
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tessa Arends
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Curtis Laborda
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, USA
| | - Jennifer R Knapp
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, USA
| | - Laura Harmacek
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, USA
| | - Brian P O'Connor
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, USA
| |
Collapse
|
19
|
Almeida ARM, Neto JL, Cachucho A, Euzébio M, Meng X, Kim R, Fernandes MB, Raposo B, Oliveira ML, Ribeiro D, Fragoso R, Zenatti PP, Soares T, de Matos MR, Corrêa JR, Duque M, Roberts KG, Gu Z, Qu C, Pereira C, Pyne S, Pyne NJ, Barreto VM, Bernard-Pierrot I, Clappier E, Mullighan CG, Grosso AR, Yunes JA, Barata JT. Interleukin-7 receptor α mutational activation can initiate precursor B-cell acute lymphoblastic leukemia. Nat Commun 2021; 12:7268. [PMID: 34907175 PMCID: PMC8671594 DOI: 10.1038/s41467-021-27197-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 11/03/2021] [Indexed: 12/13/2022] Open
Abstract
Interleukin-7 receptor α (encoded by IL7R) is essential for lymphoid development. Whether acute lymphoblastic leukemia (ALL)-related IL7R gain-of-function mutations can trigger leukemogenesis remains unclear. Here, we demonstrate that lymphoid-restricted mutant IL7R, expressed at physiological levels in conditional knock-in mice, establishes a pre-leukemic stage in which B-cell precursors display self-renewal ability, initiating leukemia resembling PAX5 P80R or Ph-like human B-ALL. Full transformation associates with transcriptional upregulation of oncogenes such as Myc or Bcl2, downregulation of tumor suppressors such as Ikzf1 or Arid2, and major IL-7R signaling upregulation (involving JAK/STAT5 and PI3K/mTOR), required for leukemia cell viability. Accordingly, maximal signaling drives full penetrance and early leukemia onset in homozygous IL7R mutant animals. Notably, we identify 2 transcriptional subgroups in mouse and human Ph-like ALL, and show that dactolisib and sphingosine-kinase inhibitors are potential treatment avenues for IL-7R-related cases. Our model, a resource to explore the pathophysiology and therapeutic vulnerabilities of B-ALL, demonstrates that IL7R can initiate this malignancy.
Collapse
Affiliation(s)
- Afonso R. M. Almeida
- grid.9983.b0000 0001 2181 4263Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João L. Neto
- grid.9983.b0000 0001 2181 4263Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Cachucho
- grid.9983.b0000 0001 2181 4263Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Mayara Euzébio
- grid.9983.b0000 0001 2181 4263Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal ,grid.456556.1Centro Infantil Boldrini, Campinas, SP Brazil
| | - Xiangyu Meng
- grid.4444.00000 0001 2112 9282Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, Paris, France
| | - Rathana Kim
- grid.413328.f0000 0001 2300 6614Hematology Laboratory, Saint-Louis Hospital, AP-HP, Paris, France, and Saint-Louis Research Institute, Université de Paris, INSERM U944/Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7212, Paris, France
| | - Marta B. Fernandes
- grid.9983.b0000 0001 2181 4263Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Beatriz Raposo
- grid.9983.b0000 0001 2181 4263Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Mariana L. Oliveira
- grid.9983.b0000 0001 2181 4263Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Daniel Ribeiro
- grid.9983.b0000 0001 2181 4263Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rita Fragoso
- grid.9983.b0000 0001 2181 4263Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | | | - Tiago Soares
- grid.9983.b0000 0001 2181 4263Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Mafalda R. de Matos
- grid.9983.b0000 0001 2181 4263Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | | | - Mafalda Duque
- grid.9983.b0000 0001 2181 4263Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Kathryn G. Roberts
- grid.240871.80000 0001 0224 711XDepartment of Pathology and Hematological Malignancies Program, St. Jude Children’s Research Hospital, Memphis, TN US
| | - Zhaohui Gu
- grid.240871.80000 0001 0224 711XDepartment of Pathology and Hematological Malignancies Program, St. Jude Children’s Research Hospital, Memphis, TN US
| | - Chunxu Qu
- grid.240871.80000 0001 0224 711XDepartment of Pathology and Hematological Malignancies Program, St. Jude Children’s Research Hospital, Memphis, TN US
| | - Clara Pereira
- grid.8217.c0000 0004 1936 9705Smurfit Institute of Genetics, Trinity College Dublin, University of Dublin, Dublin 2, Ireland
| | - Susan Pyne
- grid.11984.350000000121138138Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, Scotland UK
| | - Nigel J. Pyne
- grid.11984.350000000121138138Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, Scotland UK
| | - Vasco M. Barreto
- grid.10772.330000000121511713DNA Breaks Laboratory, CEDOC - Chronic Diseases Research Center, NOVA Medical School - Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Isabelle Bernard-Pierrot
- grid.4444.00000 0001 2112 9282Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, Paris, France
| | - Emannuelle Clappier
- grid.413328.f0000 0001 2300 6614Hematology Laboratory, Saint-Louis Hospital, AP-HP, Paris, France, and Saint-Louis Research Institute, Université de Paris, INSERM U944/Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7212, Paris, France
| | - Charles G. Mullighan
- grid.240871.80000 0001 0224 711XDepartment of Pathology and Hematological Malignancies Program, St. Jude Children’s Research Hospital, Memphis, TN US
| | - Ana R. Grosso
- grid.10772.330000000121511713UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | | | - João T. Barata
- grid.9983.b0000 0001 2181 4263Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
20
|
Hamza A, Amit J, Elizabeth L. E, Medha M. P, Michael D. C, Wendy F. L. Ion channel mediated mechanotransduction in immune cells. CURRENT OPINION IN SOLID STATE & MATERIALS SCIENCE 2021; 25:100951. [PMID: 35645593 PMCID: PMC9131931 DOI: 10.1016/j.cossms.2021.100951] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The immune system performs critical functions to defend against invading pathogens and maintain tissue homeostasis. Immune cells reside within or are recruited to a host of mechanically active tissues throughout the body and, as a result, are exposed to varying types and degrees of mechanical stimuli. Despite their abundance in such tissues, the role of mechanical stimuli in influencing immune cell function and the molecular mechanisms responsible for mechanics-mediated changes are still poorly understood. The recent emergence of mechanically-gated ion channels, particularly Piezo1, has provided an exciting avenue of research within the fields of mechanobiology and immunology. Numerous studies have identified roles for mechanically-gated ion channels in mechanotransduction within various different cell types, with a few recent studies in immune cells. These initial studies provide strong evidence that mechanically-gated ion channels play pivotal roles in regulating the immune system. In this review, we discuss characteristics of ion channel mediated force transduction, review the current techniques used to quantify and visualize ion channel activity in response to mechanical stimuli, and finally we provide an overview of recent studies examining the role of mechanically-gated ion channels in modulating immune cell function.
Collapse
Affiliation(s)
- Atcha Hamza
- Department of Biomedical Engineering, University of California Irvine, Irvine, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, Irvine, USA
| | - Jairaman Amit
- Department of Physiology and Biophysics, University of California Irvine, Irvine, USA
| | - Evans Elizabeth L.
- Department of Physiology and Biophysics, University of California Irvine, Irvine, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, USA
| | - Pathak Medha M.
- Department of Biomedical Engineering, University of California Irvine, Irvine, USA
- Department of Physiology and Biophysics, University of California Irvine, Irvine, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, USA
| | - Cahalan Michael D.
- Department of Physiology and Biophysics, University of California Irvine, Irvine, USA
| | - Liu Wendy F.
- Department of Biomedical Engineering, University of California Irvine, Irvine, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, Irvine, USA
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, USA
| |
Collapse
|
21
|
Salt inducible kinases 2 and 3 are required for thymic T cell development. Sci Rep 2021; 11:21550. [PMID: 34732767 PMCID: PMC8566462 DOI: 10.1038/s41598-021-00986-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 10/15/2021] [Indexed: 12/22/2022] Open
Abstract
Salt Inducible Kinases (SIKs), of which there are 3 isoforms, are established to play roles in innate immunity, metabolic control and neuronal function, but their role in adaptive immunity is unknown. To address this gap, we used a combination of SIK knockout and kinase-inactive knock-in mice. The combined loss of SIK1 and SIK2 activity did not block T cell development. Conditional knockout of SIK3 in haemopoietic cells, driven by a Vav-iCre transgene, resulted in a moderate reduction in the numbers of peripheral T cells, but normal B cell numbers. Constitutive knockout of SIK2 combined with conditional knockout of SIK3 in the haemopoietic cells resulted in a severe reduction in peripheral T cells without reducing B cell number. A similar effect was seen when SIK3 deletion was driven via CD4-Cre transgene to delete at the DP stage of T cell development. Analysis of the SIK2/3 Vav-iCre mice showed that thymocyte number was greatly reduced, but development was not blocked completely as indicated by the presence of low numbers CD4 and CD8 single positive cells. SIK2 and SIK3 were not required for rearrangement of the TCRβ locus, or for low level cell surface expression of the TCR complex on the surface of CD4/CD8 double positive thymocytes. In the absence of both SIK2 and SIK3, progression to mature single positive cells was greatly reduced, suggesting a defect in negative and/or positive selection in the thymus. In agreement with an effect on negative selection, increased apoptosis was seen in thymic TCRbeta high/CD5 positive cells from SIK2/3 knockout mice. Together, these results show an important role for SIK2 and SIK3 in thymic T cell development.
Collapse
|
22
|
Liu X, Yu J, Xu L, Umphred-Wilson K, Peng F, Ding Y, Barton BM, Lv X, Zhao MY, Sun S, Hong Y, Qi L, Adoro S, Chen X. Notch-induced endoplasmic reticulum-associated degradation governs mouse thymocyte β-selection. eLife 2021; 10:e69975. [PMID: 34240701 PMCID: PMC8315795 DOI: 10.7554/elife.69975] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/05/2021] [Indexed: 11/13/2022] Open
Abstract
Signals from the pre-T cell receptor and Notch coordinately instruct β-selection of CD4-CD8-double negative (DN) thymocytes to generate αβ T cells in the thymus. However, how these signals ensure a high-fidelity proteome and safeguard the clonal diversification of the pre-selection TCR repertoire given the considerable translational activity imposed by β-selection is largely unknown. Here, we identify the endoplasmic reticulum (ER)-associated degradation (ERAD) machinery as a critical proteostasis checkpoint during β-selection. Expression of the SEL1L-HRD1 complex, the most conserved branch of ERAD, is directly regulated by the transcriptional activity of the Notch intracellular domain. Deletion of Sel1l impaired DN3 to DN4 thymocyte transition and severely impaired mouse αβ T cell development. Mechanistically, Sel1l deficiency induced unresolved ER stress that triggered thymocyte apoptosis through the PERK pathway. Accordingly, genetically inactivating PERK rescued T cell development from Sel1l-deficient thymocytes. In contrast, IRE1α/XBP1 pathway was induced as a compensatory adaptation to alleviate Sel1l-deficiency-induced ER stress. Dual loss of Sel1l and Xbp1 markedly exacerbated the thymic defect. Our study reveals a critical developmental signal controlled proteostasis mechanism that enforces T cell development to ensure a healthy adaptive immunity.
Collapse
Affiliation(s)
- Xia Liu
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of MedicineHoustonUnited States
| | - Jingjing Yu
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of MedicineHoustonUnited States
| | - Longyong Xu
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of MedicineHoustonUnited States
| | - Katharine Umphred-Wilson
- Department of Pathology, School of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Fanglue Peng
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of MedicineHoustonUnited States
| | - Yao Ding
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of MedicineHoustonUnited States
| | - Brendan M Barton
- Department of Pathology, School of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Xiangdong Lv
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Michael Y Zhao
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Shengyi Sun
- Center for Molecular Medicine and Genetics, Wayne State UniversityDetroitUnited States
| | - Yuning Hong
- Department of Chemistry and Physics, La Trobe UniversityMelbourneAustralia
| | - Ling Qi
- Department of Molecular and Integrative Physiology, University of Michigan Medical SchoolAnn ArborUnited States
| | - Stanley Adoro
- Department of Pathology, School of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Xi Chen
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
23
|
Yousefzadeh MJ, Flores RR, Zhu Y, Schmiechen ZC, Brooks RW, Trussoni CE, Cui Y, Angelini L, Lee KA, McGowan SJ, Burrack AL, Wang D, Dong Q, Lu A, Sano T, O'Kelly RD, McGuckian CA, Kato JI, Bank MP, Wade EA, Pillai SPS, Klug J, Ladiges WC, Burd CE, Lewis SE, LaRusso NF, Vo NV, Wang Y, Kelley EE, Huard J, Stromnes IM, Robbins PD, Niedernhofer LJ. An aged immune system drives senescence and ageing of solid organs. Nature 2021; 594:100-105. [PMID: 33981041 PMCID: PMC8684299 DOI: 10.1038/s41586-021-03547-7] [Citation(s) in RCA: 516] [Impact Index Per Article: 129.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 04/13/2021] [Indexed: 11/09/2022]
Abstract
Ageing of the immune system, or immunosenescence, contributes to the morbidity and mortality of the elderly1,2. To define the contribution of immune system ageing to organism ageing, here we selectively deleted Ercc1, which encodes a crucial DNA repair protein3,4, in mouse haematopoietic cells to increase the burden of endogenous DNA damage and thereby senescence5-7 in the immune system only. We show that Vav-iCre+/-;Ercc1-/fl mice were healthy into adulthood, then displayed premature onset of immunosenescence characterized by attrition and senescence of specific immune cell populations and impaired immune function, similar to changes that occur during ageing in wild-type mice8-10. Notably, non-lymphoid organs also showed increased senescence and damage, which suggests that senescent, aged immune cells can promote systemic ageing. The transplantation of splenocytes from Vav-iCre+/-;Ercc1-/fl or aged wild-type mice into young mice induced senescence in trans, whereas the transplantation of young immune cells attenuated senescence. The treatment of Vav-iCre+/-;Ercc1-/fl mice with rapamycin reduced markers of senescence in immune cells and improved immune function11,12. These data demonstrate that an aged, senescent immune system has a causal role in driving systemic ageing and therefore represents a key therapeutic target to extend healthy ageing.
Collapse
Affiliation(s)
- Matthew J Yousefzadeh
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Rafael R Flores
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Zoe C Schmiechen
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Robert W Brooks
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Christy E Trussoni
- Division of Gastroenterology, Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN, USA
| | - Yuxiang Cui
- Department of Chemistry, University of California Riverside, Riverside, CA, USA
| | - Luise Angelini
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Kyoo-A Lee
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Sara J McGowan
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Adam L Burrack
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Dong Wang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qing Dong
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aiping Lu
- Department of Orthopedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tokio Sano
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Ryan D O'Kelly
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Collin A McGuckian
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Jonathan I Kato
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Michael P Bank
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Erin A Wade
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | | | - Jenna Klug
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Warren C Ladiges
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Christin E Burd
- Departments of Molecular Genetics and Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| | - Sara E Lewis
- Department of Physiology & Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Nicholas F LaRusso
- Division of Gastroenterology, Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN, USA
| | - Nam V Vo
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yinsheng Wang
- Department of Chemistry, University of California Riverside, Riverside, CA, USA
| | - Eric E Kelley
- Department of Physiology & Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Johnny Huard
- Department of Orthopedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ingunn M Stromnes
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA.
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA.
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA.
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
24
|
Royer DJ, Cook DN. Regulation of Immune Responses by Nonhematopoietic Cells in Asthma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:292-301. [PMID: 33397743 PMCID: PMC8581969 DOI: 10.4049/jimmunol.2000885] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022]
Abstract
Nonhematopoietic cells are emerging as important contributors to many inflammatory diseases, including allergic asthma. Recent advances have led to a deeper understanding of how these cells interact with traditional immune cells, thereby modulating their activities in both homeostasis and disease. In addition to their well-established roles in gas exchange and barrier function, lung epithelial cells express an armament of innate sensors that can be triggered by various inhaled environmental agents, leading to the production of proinflammatory molecules. Advances in cell lineage tracing and single-cell RNA sequencing have expanded our knowledge of rare, but immunologically important nonhematopoietic cell populations. In parallel with these advances, novel reverse genetic approaches are revealing how individual genes in different lung-resident nonhematopoietic cell populations contribute to the initiation and maintenance of asthma. This knowledge is already revealing new pathways that can be selectively targeted to treat distinct forms of asthma.
Collapse
Affiliation(s)
- Derek J Royer
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Donald N Cook
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| |
Collapse
|
25
|
Stearoyl-CoA Desaturase-Mediated Monounsaturated Fatty Acid Availability Supports Humoral Immunity. Cell Rep 2021; 34:108601. [PMID: 33406440 PMCID: PMC7839063 DOI: 10.1016/j.celrep.2020.108601] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/13/2020] [Accepted: 12/14/2020] [Indexed: 01/23/2023] Open
Abstract
Immune cells can metabolize glucose, amino acids, and fatty acids (FAs) to generate energy. The roles of different FA species and their impacts on humoral immunity remain poorly understood. Here, we report that proliferating B cells require monounsaturated FAs (MUFAs) to maintain mitochondrial metabolism and mTOR activity and to prevent excessive autophagy and endoplasmic reticulum (ER) stress. Furthermore, B cell-extrinsic stearoyl-CoA desaturase (SCD) activity generates MUFA to support early B cell development and germinal center (GC) formation in vivo during immunization and influenza infection. Thus, SCD-mediated MUFA production is critical for humoral immunity. Zhou et al. show that monounsaturated fatty acids (MUFAs), generated by stearoyl-CoA desaturase (SCD), support B cell mitochondrial metabolism and mTOR activity and promote B cell development and humoral immune responses. These data establish MUFA availability as a key regulator for humoral immunity and a potential therapeutic target.
Collapse
|
26
|
Köchl R, Vanes L, Llorian Sopena M, Chakravarty P, Hartweger H, Fountain K, White A, Cowan J, Anderson G, Tybulewicz VLJ. Critical role of WNK1 in MYC-dependent early mouse thymocyte development. eLife 2020; 9:e56934. [PMID: 33051000 PMCID: PMC7591260 DOI: 10.7554/elife.56934] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 10/13/2020] [Indexed: 11/13/2022] Open
Abstract
WNK1, a kinase that controls kidney salt homeostasis, also regulates adhesion and migration in CD4+ T cells. Wnk1 is highly expressed in thymocytes, and since migration is important for thymocyte maturation, we investigated a role for WNK1 in mouse thymocyte development. We find that WNK1 is required for the transition of double negative (DN) thymocytes through the β-selection checkpoint and subsequent proliferation and differentiation into double positive (DP) thymocytes. Furthermore, we show that WNK1 negatively regulates LFA1-mediated adhesion and positively regulates CXCL12-induced migration in DN thymocytes. Despite this, migration defects of WNK1-deficient thymocytes do not account for the developmental arrest. Instead, we show that in DN thymocytes WNK1 transduces pre-TCR signals via OXSR1 and STK39 kinases, and the SLC12A2 ion co-transporter that are required for post-transcriptional upregulation of MYC and subsequent proliferation and differentiation into DP thymocytes. Thus, a pathway regulating ion homeostasis is a critical regulator of thymocyte development.
Collapse
Affiliation(s)
- Robert Köchl
- The Francis Crick InstituteLondonUnited Kingdom
- Kings College LondonLondonUnited Kingdom
| | | | | | | | | | | | - Andrea White
- University of BirminghamBirminghamUnited Kingdom
| | | | | | - Victor LJ Tybulewicz
- The Francis Crick InstituteLondonUnited Kingdom
- Imperial CollegeLondonUnited Kingdom
| |
Collapse
|
27
|
Astori A, Tingvall-Gustafsson J, Kuruvilla J, Coyaud E, Laurent EMN, Sunnerhagen M, Åhsberg J, Ungerbäck J, Strid T, Sigvardsson M, Raught B, Somasundaram R. ARID1a Associates with Lymphoid-Restricted Transcription Factors and Has an Essential Role in T Cell Development. THE JOURNAL OF IMMUNOLOGY 2020; 205:1419-1432. [PMID: 32747500 DOI: 10.4049/jimmunol.1900959] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 06/29/2020] [Indexed: 11/19/2022]
Abstract
Maturation of lymphoid cells is controlled by the action of stage and lineage-restricted transcription factors working in concert with the general transcription and chromatin remodeling machinery to regulate gene expression. To better understand this functional interplay, we used Biotin Identification in human embryonic kidney cells to identify proximity interaction partners for GATA3, TCF7 (TCF1), SPI1, HLF, IKZF1, PAX5, ID1, and ID2. The proximity interaction partners shared among the lineage-restricted transcription factors included ARID1a, a BRG1-associated factor complex component. CUT&RUN analysis revealed that ARID1a shared binding with TCF7 and GATA3 at a substantial number of putative regulatory elements in mouse T cell progenitors. In support of an important function for ARID1a in lymphocyte development, deletion of Arid1a in early lymphoid progenitors in mice resulted in a pronounced developmental arrest in early T cell development with a reduction of CD4+CD8+ cells and a 20-fold reduction in thymic cellularity. Exploring gene expression patterns in DN3 cells from Wt and Arid1a-deficient mice suggested that the developmental block resided in the DN3a to DN3b transition, indicating a deficiency in β-selection. Our work highlights the critical importance of functional interactions between stage and lineage-restricted factors and the basic transcription machinery during lymphocyte differentiation.
Collapse
Affiliation(s)
- Audrey Astori
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | | | - Jacob Kuruvilla
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Etienne Coyaud
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Estelle M N Laurent
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Maria Sunnerhagen
- Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden; and
| | - Josefine Åhsberg
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Jonas Ungerbäck
- Division of Molecular Hematology, Lund University, 22184 Lund, Sweden
| | - Tobias Strid
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Mikael Sigvardsson
- Division of Molecular Hematology, Lund University, 22184 Lund, Sweden; .,Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5S 3K1, Canada
| | - Rajesh Somasundaram
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| |
Collapse
|
28
|
Chu SH, Chabon JR, Matovina CN, Minehart JC, Chen BR, Zhang J, Kumar V, Xiong Y, Callen E, Hung PJ, Feng Z, Koche RP, Liu XS, Chaudhuri J, Nussenzweig A, Sleckman BP, Armstrong SA. Loss of H3K36 Methyltransferase SETD2 Impairs V(D)J Recombination during Lymphoid Development. iScience 2020; 23:100941. [PMID: 32169821 PMCID: PMC7066224 DOI: 10.1016/j.isci.2020.100941] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/25/2020] [Accepted: 02/21/2020] [Indexed: 12/17/2022] Open
Abstract
Repair of DNA double-stranded breaks (DSBs) during lymphocyte development is essential for V(D)J recombination and forms the basis of immunoglobulin variable region diversity. Understanding of this process in lymphogenesis has historically been centered on the study of RAG1/2 recombinases and a set of classical non-homologous end-joining factors. Much less has been reported regarding the role of chromatin modifications on this process. Here, we show a role for the non-redundant histone H3 lysine methyltransferase, Setd2, and its modification of lysine-36 trimethylation (H3K36me3), in the processing and joining of DNA ends during V(D)J recombination. Loss leads to mis-repair of Rag-induced DNA DSBs, especially when combined with loss of Atm kinase activity. Furthermore, loss reduces immune repertoire and a severe block in lymphogenesis as well as causes post-mitotic neuronal apoptosis. Together, these studies are suggestive of an important role of Setd2/H3K36me3 in these two mammalian developmental processes that are influenced by double-stranded break repair.
Collapse
Affiliation(s)
- S Haihua Chu
- Department of Pediatric Oncology, Dana Farber Cancer Institute, and Division of Hematology/Oncology, Boston Children's Hospital, 450 Brookline Avenue, Boston, MA 02215-5450, USA
| | - Jonathan R Chabon
- Department of Pediatric Oncology, Dana Farber Cancer Institute, and Division of Hematology/Oncology, Boston Children's Hospital, 450 Brookline Avenue, Boston, MA 02215-5450, USA
| | - Chloe N Matovina
- Department of Pediatric Oncology, Dana Farber Cancer Institute, and Division of Hematology/Oncology, Boston Children's Hospital, 450 Brookline Avenue, Boston, MA 02215-5450, USA
| | | | - Bo-Ruei Chen
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Jian Zhang
- Center for Computational Biology, Beijing Institute of Basic Medical Sciences, Beijing, China; Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Vipul Kumar
- Howard Hughes Medical Institute, Department of Pediatrics, Department of Genetics, Harvard Medical School, Boston, MA, USA; Harvard-MIT MD-PhD Program, Harvard Medical School, Boston, MA, USA
| | - Yijun Xiong
- Department of Pediatric Oncology, Dana Farber Cancer Institute, and Division of Hematology/Oncology, Boston Children's Hospital, 450 Brookline Avenue, Boston, MA 02215-5450, USA
| | - Elsa Callen
- Laboratory of Genome Integrity, National Cancer Institute National Institutes of Health, Bethesda, MD, USA
| | - Putzer J Hung
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Zhaohui Feng
- Department of Pediatric Oncology, Dana Farber Cancer Institute, and Division of Hematology/Oncology, Boston Children's Hospital, 450 Brookline Avenue, Boston, MA 02215-5450, USA
| | - Richard P Koche
- Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - X Shirley Liu
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jayanta Chaudhuri
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Andre Nussenzweig
- Laboratory of Genome Integrity, National Cancer Institute National Institutes of Health, Bethesda, MD, USA
| | - Barry P Sleckman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Scott A Armstrong
- Department of Pediatric Oncology, Dana Farber Cancer Institute, and Division of Hematology/Oncology, Boston Children's Hospital, 450 Brookline Avenue, Boston, MA 02215-5450, USA.
| |
Collapse
|
29
|
Agerholm R, Rizk J, Viñals MT, Bekiaris V. STAT3 but not STAT4 is critical for γδT17 cell responses and skin inflammation. EMBO Rep 2019; 20:e48647. [PMID: 31549795 PMCID: PMC6832010 DOI: 10.15252/embr.201948647] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/23/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022] Open
Abstract
The transcription factors STAT3 and STAT4 are essential for lymphocyte differentiation and function. Interleukin (IL)-17 producing γδ T (γδT17) cells are innate lymphocytes important for anti-bacterial and inflammatory responses at barrier surfaces. Herein, we examine the role of STAT3 and STAT4 in regulating the homeostasis, activation, and pathogenicity of γδT17 cells. We show that STAT3 sustains γδT17 numbers in the skin but not in the lymph nodes, while STAT4 deficiency does not affect their homeostasis. Similarly, STAT3 but not STAT4 is essential for IL-23-induced IL-22 production by γδT17 cells. Concomitantly, mice lacking STAT3 expression in γδT17 cells develop significantly reduced psoriasis-like inflammation. STAT3-deficient γδT17 cells fail to expand and to upregulate IL-17A, IL-17F, and IL-22 in response to psoriatic stimuli. Although STAT4-deficient animals develop psoriasis-like disease, γδT17 cells in these mice are defective in IL-17F production. Collectively, our data demonstrate for the first time a critical role for STAT3 in orchestrating the homeostasis and pathogenicity of γδT17 cells and provide evidence for the requirement of STAT4 for optimal cytokine responses during inflammation.
Collapse
Affiliation(s)
- Rasmus Agerholm
- Department of Health TechnologyTechnical University of DenmarkKgs LyngbyDenmark
| | - John Rizk
- Department of Health TechnologyTechnical University of DenmarkKgs LyngbyDenmark
| | | | - Vasileios Bekiaris
- Department of Health TechnologyTechnical University of DenmarkKgs LyngbyDenmark
| |
Collapse
|
30
|
Peterson TV, Jaiswal MK, Beaman KD, Reynolds JM. Conditional Deletion of the V-ATPase a2-Subunit Disrupts Intrathymic T Cell Development. Front Immunol 2019; 10:1911. [PMID: 31456807 PMCID: PMC6700305 DOI: 10.3389/fimmu.2019.01911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 07/29/2019] [Indexed: 11/13/2022] Open
Abstract
Proper orchestration of T lymphocyte development is critical, as T cells underlie nearly all responses of the adaptive immune system. Developing thymocytes differentiate in response to environmental cues carried from cell surface receptors to the nucleus, shaping a distinct transcriptional program that defines their developmental outcome. Our recent work has identified a previously undescribed role for the vacuolar ATPase (V-ATPase) in facilitating the development of murine thymocytes progressing toward the CD4+ and CD8+ αβ T cell lineages. Vav1Cre recombinase-mediated deletion of the a2 isoform of the V-ATPase (a2V) in mouse hematopoietic cells leads to a specific and profound loss of peripheral CD4+ and CD8+ αβ T cells. Utilizing T cell-restricted LckCre and CD4Cre strains, we further traced this deficiency to the thymus and found that a2V plays a cell-intrinsic role throughout intrathymic development. Loss of a2V manifests as a partial obstruction in the double negative stage of T cell development, and later, a near complete failure of positive selection. These data deepen our understanding of the biological mechanisms that orchestrate T cell development and lend credence to the recent focus on V-ATPase as a potential chemotherapeutic target to combat proliferative potential in T cell lymphoblastic leukemias and autoimmune disease.
Collapse
Affiliation(s)
- Theodore V Peterson
- Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Mukesh K Jaiswal
- Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Kenneth D Beaman
- Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Joseph M Reynolds
- Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Edward Hines, Jr. VA Hospital, Hines, IL, United States
| |
Collapse
|
31
|
Colijn S, Gao S, Ingram KG, Menendez M, Muthukumar V, Silasi-Mansat R, Chmielewska JJ, Hinsdale M, Lupu F, Griffin CT. The NuRD chromatin-remodeling complex enzyme CHD4 prevents hypoxia-induced endothelial Ripk3 transcription and murine embryonic vascular rupture. Cell Death Differ 2019; 27:618-631. [PMID: 31235857 DOI: 10.1038/s41418-019-0376-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 05/10/2019] [Accepted: 06/11/2019] [Indexed: 01/06/2023] Open
Abstract
Physiological hypoxia can trigger transcriptional events that influence many developmental processes during mammalian embryogenesis. One way that hypoxia affects transcription is by engaging chromatin-remodeling complexes. We now report that chromodomain helicase DNA binding protein 4 (CHD4), an enzyme belonging to the nucleosome remodeling and deacetylase (NuRD) chromatin-remodeling complex, is required for transcriptional repression of the receptor-interacting protein kinase 3 (Ripk3)-a critical executor of the necroptosis cell death program-in hypoxic murine embryonic endothelial cells. Genetic deletion of Chd4 in murine embryonic endothelial cells in vivo results in upregulation of Ripk3 transcripts and protein prior to vascular rupture and lethality at midgestation, and concomitant deletion of Ripk3 partially rescues these phenotypes. In addition, CHD4 binds to and prevents acetylation of the Ripk3 promoter in cultured endothelial cells grown under hypoxic conditions to prevent excessive Ripk3 transcription. These data demonstrate that excessive RIPK3 is detrimental to embryonic vascular integrity and indicate that CHD4 suppresses Ripk3 transcription when the embryonic environment is particularly hypoxic prior to the establishment of fetal-placental circulation at midgestation. Altogether, this research provides new insights into regulators of Ripk3 transcription and encourages future studies into the mechanism by which excessive RIPK3 damages embryonic blood vessels.
Collapse
Affiliation(s)
- Sarah Colijn
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73190, USA
| | - Siqi Gao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73190, USA
| | - Kyle G Ingram
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73190, USA.,Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Matthew Menendez
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Vijay Muthukumar
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.,The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | - Robert Silasi-Mansat
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Joanna J Chmielewska
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.,Centre of New Technologies, University of Warsaw, 02-097, Warsaw, Poland
| | - Myron Hinsdale
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73190, USA
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA. .,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73190, USA.
| |
Collapse
|
32
|
Dress RJ, Dutertre CA, Giladi A, Schlitzer A, Low I, Shadan NB, Tay A, Lum J, Kairi MFBM, Hwang YY, Becht E, Cheng Y, Chevrier M, Larbi A, Newell EW, Amit I, Chen J, Ginhoux F. Plasmacytoid dendritic cells develop from Ly6D+ lymphoid progenitors distinct from the myeloid lineage. Nat Immunol 2019; 20:852-864. [DOI: 10.1038/s41590-019-0420-3] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 05/13/2019] [Indexed: 01/19/2023]
|
33
|
Fiala GJ, Schaffer AM, Merches K, Morath A, Swann J, Herr LA, Hils M, Esser C, Minguet S, Schamel WWA. Proximal Lck Promoter–Driven Cre Function Is Limited in Neonatal and Ineffective in Adult γδ T Cell Development. THE JOURNAL OF IMMUNOLOGY 2019; 203:569-579. [DOI: 10.4049/jimmunol.1701521] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 05/08/2019] [Indexed: 01/13/2023]
|
34
|
Zikmund T, Kokavec J, Turkova T, Savvulidi F, Paszekova H, Vodenkova S, Sedlacek R, Skoultchi AI, Stopka T. ISWI ATPase Smarca5 Regulates Differentiation of Thymocytes Undergoing β-Selection. THE JOURNAL OF IMMUNOLOGY 2019; 202:3434-3446. [PMID: 31068388 DOI: 10.4049/jimmunol.1801684] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/15/2019] [Indexed: 01/13/2023]
Abstract
Development of lymphoid progenitors requires a coordinated regulation of gene expression, DNA replication, and gene rearrangement. Chromatin-remodeling activities directed by SWI/SNF2 superfamily complexes play important roles in these processes. In this study, we used a conditional knockout mouse model to investigate the role of Smarca5, a member of the ISWI subfamily of such complexes, in early lymphocyte development. Smarca5 deficiency results in a developmental block at the DN3 stage of αβ thymocytes and pro-B stage of early B cells at which the rearrangement of Ag receptor loci occurs. It also disturbs the development of committed (CD73+) γδ thymocytes. The αβ thymocyte block is accompanied by massive apoptotic depletion of β-selected double-negative DN3 cells and premitotic arrest of CD4/CD8 double-positive cells. Although Smarca5-deficient αβ T cell precursors that survived apoptosis were able to undergo a successful TCRβ rearrangement, they exhibited a highly abnormal mRNA profile, including the persistent expression of CD44 and CD25 markers characteristic of immature cells. We also observed that the p53 pathway became activated in these cells and that a deficiency of p53 partially rescued the defect in thymus cellularity (in contrast to early B cells) of Smarca5-deficient mice. However, the activation of p53 was not primarily responsible for the thymocyte developmental defects observed in the Smarca5 mutants. Our results indicate that Smarca5 plays a key role in the development of thymocytes undergoing β-selection, γδ thymocytes, and also B cell progenitors by regulating the transcription of early differentiation programs.
Collapse
Affiliation(s)
- Tomas Zikmund
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 25250, Czech Republic
| | - Juraj Kokavec
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 25250, Czech Republic
| | - Tereza Turkova
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 25250, Czech Republic
| | - Filipp Savvulidi
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague 12853, Czech Republic
| | - Helena Paszekova
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 25250, Czech Republic
| | - Sona Vodenkova
- Institute of Experimental Medicine, Czech Academy of Sciences, Prague 14220, Czech Republic.,Third Faculty of Medicine, Charles University, Prague 10000, Czech Republic
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics, Czech Academy of Sciences, Vestec 25250, Czech Republic; and
| | - Arthur I Skoultchi
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx 10461, NY
| | - Tomas Stopka
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 25250, Czech Republic;
| |
Collapse
|
35
|
Asrij/OCIAD1 suppresses CSN5-mediated p53 degradation and maintains mouse hematopoietic stem cell quiescence. Blood 2019; 133:2385-2400. [PMID: 30952670 DOI: 10.1182/blood.2019000530] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/18/2019] [Indexed: 12/11/2022] Open
Abstract
Inactivation of the tumor suppressor p53 is essential for unrestrained growth of cancers. However, only 11% of hematological malignancies have mutant p53. Mechanisms that cause wild-type p53 dysfunction and promote leukemia are inadequately deciphered. The stem cell protein Asrij/OCIAD1 is misexpressed in several human hematological malignancies and implicated in the p53 pathway and DNA damage response. However, Asrij function in vertebrate hematopoiesis remains unknown. We generated the first asrij null (knockout [KO]) mice and show that they are viable and fertile with no gross abnormalities. However, by 6 months, they exhibit increased peripheral blood cell counts, splenomegaly, and an expansion of bone marrow hematopoietic stem cells (HSCs) with higher myeloid output. HSCs lacking Asrij are less quiescent and more proliferative with higher repopulation potential as observed from serial transplantation studies. However, stressing KO mice with sublethal γ irradiation or multiple injections of 5-fluorouracil results in reduced survival and rapid depletion of hematopoietic stem/progenitor cells (HSPCs) by driving them into proliferative exhaustion. Molecular and biochemical analyses revealed increased polyubiquitinated protein levels, Akt/STAT5 activation and COP9 signalosome subunit 5 (CSN5)-mediated p53 ubiquitination, and degradation in KO HSPCs. Further, we show that Asrij sequesters CSN5 via its conserved OCIA domain, thereby preventing p53 degradation. In agreement, Nutlin-3 treatment of KO mice restored p53 levels and reduced high HSPC frequencies. Thus, we provide a new mouse model resembling myeloproliferative disease and identify a posttranslational regulator of wild-type p53 essential for maintaining HSC quiescence that could be a potential target for pharmacological intervention.
Collapse
|
36
|
Lu Y, Zhong MC, Qian J, Calderon V, Cruz Tleugabulova M, Mallevaey T, Veillette A. SLAM receptors foster iNKT cell development by reducing TCR signal strength after positive selection. Nat Immunol 2019; 20:447-457. [DOI: 10.1038/s41590-019-0334-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/25/2019] [Indexed: 12/23/2022]
|
37
|
de Bock CE, Demeyer S, Degryse S, Verbeke D, Sweron B, Gielen O, Vandepoel R, Vicente C, Vanden Bempt M, Dagklis A, Geerdens E, Bornschein S, Gijsbers R, Soulier J, Meijerink JP, Heinäniemi M, Teppo S, Bouvy-Liivrand M, Lohi O, Radaelli E, Cools J. HOXA9 Cooperates with Activated JAK/STAT Signaling to Drive Leukemia Development. Cancer Discov 2018; 8:616-631. [PMID: 29496663 DOI: 10.1158/2159-8290.cd-17-0583] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 01/26/2018] [Accepted: 02/22/2018] [Indexed: 11/16/2022]
Abstract
Leukemia is caused by the accumulation of multiple genomic lesions in hematopoietic precursor cells. However, how these events cooperate during oncogenic transformation remains poorly understood. We studied the cooperation between activated JAK3/STAT5 signaling and HOXA9 overexpression, two events identified as significantly co-occurring in T-cell acute lymphoblastic leukemia. Expression of mutant JAK3 and HOXA9 led to a rapid development of leukemia originating from multipotent or lymphoid-committed progenitors, with a significant decrease in disease latency compared with JAK3 or HOXA9 alone. Integrated RNA sequencing, chromatin immunoprecipitation sequencing, and Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) revealed that STAT5 and HOXA9 have co-occupancy across the genome, resulting in enhanced STAT5 transcriptional activity and ectopic activation of FOS/JUN (AP1). Our data suggest that oncogenic transcription factors such as HOXA9 provide a fertile ground for specific signaling pathways to thrive, explaining why JAK/STAT pathway mutations accumulate in HOXA9-expressing cells.Significance: The mechanism of oncogene cooperation in cancer development remains poorly characterized. In this study, we model the cooperation between activated JAK/STAT signaling and ectopic HOXA9 expression during T-cell leukemia development. We identify a direct cooperation between STAT5 and HOXA9 at the transcriptional level and identify PIM1 kinase as a possible drug target in mutant JAK/STAT/HOXA9-positive leukemia cases. Cancer Discov; 8(5); 616-31. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 517.
Collapse
Affiliation(s)
- Charles E de Bock
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Sofie Demeyer
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Sandrine Degryse
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Delphine Verbeke
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Bram Sweron
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Olga Gielen
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Roel Vandepoel
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Carmen Vicente
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Marlies Vanden Bempt
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Antonis Dagklis
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Ellen Geerdens
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Simon Bornschein
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Rik Gijsbers
- Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Jean Soulier
- U944 INSERM and Hematology Laboratory, St-Louis Hospital, APHP, Hematology University Institute, University Paris-Diderot, Paris, France
| | - Jules P Meijerink
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Merja Heinäniemi
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Susanna Teppo
- Tampere Centre for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Maria Bouvy-Liivrand
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Olli Lohi
- Tampere Centre for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Enrico Radaelli
- KU Leuven, Center for Human Genetics, Leuven, Belgium.,VIB, Center for Cancer Biology, Leuven, Belgium
| | - Jan Cools
- KU Leuven, Center for Human Genetics, Leuven, Belgium. .,VIB, Center for Cancer Biology, Leuven, Belgium
| |
Collapse
|
38
|
Zeng H, Yu M, Tan H, Li Y, Su W, Shi H, Dhungana Y, Guy C, Neale G, Cloer C, Peng J, Wang D, Chi H. Discrete roles and bifurcation of PTEN signaling and mTORC1-mediated anabolic metabolism underlie IL-7-driven B lymphopoiesis. SCIENCE ADVANCES 2018; 4:eaar5701. [PMID: 29399633 PMCID: PMC5792226 DOI: 10.1126/sciadv.aar5701] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/04/2018] [Indexed: 05/21/2023]
Abstract
Interleukin-7 (IL-7) drives early B lymphopoiesis, but the underlying molecular circuits remain poorly understood, especially how Stat5 (signal transducer and activator of transcription 5)-dependent and Stat5-independent pathways contribute to this process. Combining transcriptome and proteome analyses and mouse genetic models, we show that IL-7 promotes anabolic metabolism and biosynthetic programs in pro-B cells. IL-7-mediated activation of mTORC1 (mechanistic target of rapamycin complex 1) supported cell proliferation and metabolism in a Stat5-independent, Myc-dependent manner but was largely dispensable for cell survival or Rag1 and Rag2 gene expression. mTORC1 was also required for Myc-driven lymphomagenesis. PI3K (phosphatidylinositol 3-kinase) and mTORC1 had discrete effects on Stat5 signaling and independently controlled B cell development. PI3K was actively suppressed by PTEN (phosphatase and tensin homolog) in pro-B cells to ensure proper IL-7R expression, Stat5 activation, heavy chain rearrangement, and cell survival, suggesting the unexpected bifurcation of the classical PI3K-mTOR signaling. Together, our integrative analyses establish IL-7R-mTORC1-Myc and PTEN-mediated PI3K suppression as discrete signaling axes driving B cell development, with differential effects on IL-7R-Stat5 signaling.
Collapse
Affiliation(s)
- Hu Zeng
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Mei Yu
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI 53226, USA
| | - Haiyan Tan
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- St. Jude Proteomics Facility, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Yuxin Li
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- St. Jude Proteomics Facility, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Wei Su
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Hao Shi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Yogesh Dhungana
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Cliff Guy
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Geoffrey Neale
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Caryn Cloer
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- St. Jude Proteomics Facility, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Corresponding author. (H.C.); (D.W.); (J.P.)
| | - Demin Wang
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI 53226, USA
- Corresponding author. (H.C.); (D.W.); (J.P.)
| | - Hongbo Chi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Corresponding author. (H.C.); (D.W.); (J.P.)
| |
Collapse
|
39
|
The podoplanin-CLEC-2 axis inhibits inflammation in sepsis. Nat Commun 2017; 8:2239. [PMID: 29269852 PMCID: PMC5740111 DOI: 10.1038/s41467-017-02402-6] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 11/28/2017] [Indexed: 12/26/2022] Open
Abstract
Platelets play a critical role in vascular inflammation through the podoplanin and collagen/fibrin receptors, C-type-lectin-like-2 (CLEC-2) and glycoprotein VI (GPVI), respectively. Both receptors regulate endothelial permeability and prevent peri-vascular bleeding in inflammation. Here we show that platelet-specific deletion of CLEC-2 but not GPVI leads to enhanced systemic inflammation and accelerated organ injury in two mouse models of sepsis-intra-peritoneal lipopolysaccharide and cecal ligation and puncture. CLEC-2 deficiency is associated with reduced numbers of podoplanin-expressing macrophages despite increased cytokine and chemokine levels in the infected peritoneum. Pharmacological inhibition of the interaction between CLEC-2 and podoplanin regulates immune cell infiltration and the inflammatory reaction during sepsis, suggesting that activation of podoplanin underlies the anti-inflammatory action of platelet CLEC-2. We suggest podoplanin-CLEC-2 as a novel anti-inflammatory axis regulating immune cell recruitment and activation in sepsis.
Collapse
|
40
|
Gamma Interferon Mediates Experimental Cerebral Malaria by Signaling within Both the Hematopoietic and Nonhematopoietic Compartments. Infect Immun 2017; 85:IAI.01035-16. [PMID: 28874445 PMCID: PMC5649021 DOI: 10.1128/iai.01035-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 08/23/2017] [Indexed: 12/18/2022] Open
Abstract
Experimental cerebral malaria (ECM) is a gamma interferon (IFN-γ)-dependent syndrome. However, whether IFN-γ promotes ECM through direct and synergistic targeting of multiple cell populations or by acting primarily on a specific responsive cell type is currently unknown. Here, using a panel of cell- and compartment-specific IFN-γ receptor 2 (IFN-γR2)-deficient mice, we show that IFN-γ causes ECM by signaling within both the hematopoietic and nonhematopoietic compartments. Mechanistically, hematopoietic and nonhematopoietic compartment-specific IFN-γR signaling exerts additive effects in orchestrating intracerebral inflammation, leading to the development of ECM. Surprisingly, mice with specific deletion of IFN-γR2 expression on myeloid cells, T cells, or neurons were completely susceptible to terminal ECM. Utilizing a reductionist in vitro system, we show that synergistic IFN-γ and tumor necrosis factor (TNF) stimulation promotes strong activation of brain blood vessel endothelial cells. Combined, our data show that within the hematopoietic compartment, IFN-γ causes ECM by acting redundantly or by targeting non-T cell or non-myeloid cell populations. Within the nonhematopoietic compartment, brain endothelial cells, but not neurons, may be the major target of IFN-γ leading to ECM development. Collectively, our data provide information on how IFN-γ mediates the development of cerebral pathology during malaria infection.
Collapse
|
41
|
TAK1 regulates resident macrophages by protecting lysosomal integrity. Cell Death Dis 2017; 8:e2598. [PMID: 28182011 PMCID: PMC5386472 DOI: 10.1038/cddis.2017.23] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/04/2017] [Indexed: 12/27/2022]
Abstract
Hematopoietic cell survival and death is critical for development of a functional immune system. Here, we report that a protein kinase, TAK1, is selectively required for resident macrophage integrity during embryogenesis. Hematopoietic lineage-specific deletion of Tak1 gene (Tak1HKO) caused accumulation of cellular debris in the thymus in perinatal mice. Although no overt alteration in thymocytes and blood myeloid populations was observed in Tak1HKO mice, we found that thymic and lung macrophages were diminished. In the in vitro setting, Tak1 deficiency caused profound disruption of lysosomes and killed bone marrow-derived macrophages (BMDMs) without any exogenous stressors. Inhibition of the lysosomal protease, cathepsin B, partially blocked Tak1-deficient BMDM death, suggesting that leakage of the lysosomal contents is in part the cause of cell death. To identify the trigger of this cell death, we examined involvement of TNF and Toll-like receptor pathways. Among them, we found that deletion of Tnfr1 partially rescued cell death. Finally, we show that Tnfr1 deletion partially restored thymic and lung macrophages in vivo. These results suggest that autocrine and potentially paracrine TNF kills Tak1-deficient macrophages during development. Our results reveal that TAK1 signaling maintains proper macrophage populations through protecting lysosomal integrity.
Collapse
|
42
|
Porter SN, Cluster AS, Yang W, Busken KA, Patel RM, Ryoo J, Magee JA. Fetal and neonatal hematopoietic progenitors are functionally and transcriptionally resistant to Flt3-ITD mutations. eLife 2016; 5. [PMID: 27879203 PMCID: PMC5153248 DOI: 10.7554/elife.18882] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 11/21/2016] [Indexed: 12/24/2022] Open
Abstract
The FLT3 Internal Tandem Duplication (FLT3ITD) mutation is common in adult acute myeloid leukemia (AML) but rare in early childhood AML. It is not clear why this difference occurs. Here we show that Flt3ITD and cooperating Flt3ITD/Runx1 mutations cause hematopoietic stem cell depletion and myeloid progenitor expansion during adult but not fetal stages of murine development. In adult progenitors, FLT3ITD simultaneously induces self-renewal and myeloid commitment programs via STAT5-dependent and STAT5-independent mechanisms, respectively. While FLT3ITD can activate STAT5 signal transduction prior to birth, this signaling does not alter gene expression until hematopoietic progenitors transition from fetal to adult transcriptional states. Cooperative interactions between Flt3ITD and Runx1 mutations are also blunted in fetal/neonatal progenitors. Fetal/neonatal progenitors may therefore be protected from leukemic transformation because they are not competent to express FLT3ITD target genes. Changes in the transcriptional states of developing hematopoietic progenitors may generally shape the mutation spectra of human leukemias. DOI:http://dx.doi.org/10.7554/eLife.18882.001 Leukemias are a group of blood cancers that usually arise when immature blood cells gain one or more tumor-promoting genetic mutations. However, for reasons that are not clear, the mutations that cause leukemia are different in children and adults. For example, a mutation called FLT3ITD occurs relatively often in adult leukemia but is rare in infant leukemia. This raises the question of whether the blood cells of fetuses and babies are somehow protected from the effects of the mutation. Porter et al. have now compared the effects of the FLT3ITDmutation in blood cells from adult and fetal mice. In adult mice, the FLT3ITD mutation caused immature blood cells to turn different genes on and off. By contrast, the mutation had no effect on the activity of these genes in fetal mice. Furthermore, only the adult mutant cells showed changes that indicated the early stages of leukemia: the mutant blood cells of fetuses developed as normal. Porter et al. therefore concluded that the immature blood cells of fetuses are protected from the FLT3ITDmutation. To understand why fetal and adult blood cells respond differently to the FLT3ITDmutation, further experiments are needed to investigate how various genes regulate normal blood cell development. In addition, understanding why adult blood cells react to the FLT3ITDmutation might, in the future, lead to better treatment options for leukemia. DOI:http://dx.doi.org/10.7554/eLife.18882.002
Collapse
Affiliation(s)
- Shaina N Porter
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, United States
| | - Andrew S Cluster
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, United States
| | - Wei Yang
- Department of Genetics, Washington University School of Medicine, St. Louis, United States
| | - Kelsey A Busken
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, United States
| | - Riddhi M Patel
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, United States
| | - Jiyeon Ryoo
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, United States
| | - Jeffrey A Magee
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, United States.,Department of Genetics, Washington University School of Medicine, St. Louis, United States
| |
Collapse
|
43
|
Philips RL, Chen MW, McWilliams DC, Belmonte PJ, Constans MM, Shapiro VS. HDAC3 Is Required for the Downregulation of RORγt during Thymocyte Positive Selection. THE JOURNAL OF IMMUNOLOGY 2016; 197:541-54. [PMID: 27279370 DOI: 10.4049/jimmunol.1502529] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/09/2016] [Indexed: 12/31/2022]
Abstract
To generate functional peripheral T cells, proper gene regulation during T cell development is critical. In this study, we found that histone deacetylase (HDAC) 3 is required for T cell development. T cell development in CD2-icre HDAC3 conditional knockout (cKO) mice (HDAC3-cKO) was blocked at positive selection, resulting in few CD4 and CD8 T cells, and it could not be rescued by a TCR transgene. These single-positive thymocytes failed to upregulate Bcl-2, leading to increased apoptosis. HDAC3-cKO mice failed to downregulate retinoic acid-related orphan receptor (ROR) γt during positive selection, similar to the block in positive selection in RORγt transgenic mice. In the absence of HDAC3, the RORC promoter was hyperacetylated. In the periphery, the few CD4 T cells present were skewed toward RORγt(+) IL-17-producing Th17 cells, leading to inflammatory bowel disease. Positive selection of CD8 single-positive thymocytes was restored in RORγt-KO Bcl-xL transgenic HDAC3-cKO mice, demonstrating that HDAC3 is required at positive selection to downregulate RORγt.
Collapse
Affiliation(s)
| | - Meibo W Chen
- Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | | | | | | | | |
Collapse
|
44
|
Abdulaal WH, Walker CR, Costello R, Redondo‐Castro E, Mufazalov IA, Papaemmanouil A, Rothwell NJ, Allan SM, Waisman A, Pinteaux E, Müller W. Characterization of a conditional interleukin-1 receptor 1 mouse mutant using the Cre/LoxP system. Eur J Immunol 2016; 46:912-8. [PMID: 26692072 PMCID: PMC4982085 DOI: 10.1002/eji.201546075] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/22/2015] [Accepted: 12/15/2015] [Indexed: 11/05/2022]
Abstract
IL-1 is a key cytokine known to drive chronic inflammation and to regulate many physiological, immunological, and neuroimmunological responses via actions on diverse cell types of the body. To determine the mechanisms of IL-1 actions as part of the inflammatory response in vivo, we generated a conditional IL-1 receptor 1 (IL-1R1) mouse mutant using the Cre/LoxP system (IL-1R1(fl/fl) ). In the mutant generated, exon 5, which encodes part of the extracellular-binding region of the receptor, is flanked by LoxP sites, thereby inactivating the two previously described functional IL-1R1 gene transcripts after Cre-mediated recombination. Using keratin 14-Cre driver mice, new IL-1R1 deficient (-/-) mice were subsequently generated, in which all signaling IL-1 receptor isoforms are deleted ubiquitously. Furthermore, using vav-iCre driver mice, we deleted IL-1 receptor isoforms in the hematopoietic system. In these mice, we show that both the IL-17 and IL-22 cytokine response is reduced, when mice are challenged by the helminth Trichuris muris. We are currently crossing IL-1R1(fl/fl) mice with different Cre-expressing mice in order to study mechanisms of acute and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Wesam H. Abdulaal
- Faculty of Life SciencesUniversity of ManchesterManchesterUK
- Biochemistry Department, Faculty of SciencesKing Abdulaziz UniversityJeddahKingdom of Saudi Arabia
| | | | - Ryan Costello
- Faculty of Life SciencesUniversity of ManchesterManchesterUK
| | | | - Ilgiz A. Mufazalov
- Institute for Molecular MedicineMedical University of Johannes Gutenberg‐University of MainzMainzGermany
| | | | | | - Stuart M. Allan
- Faculty of Life SciencesUniversity of ManchesterManchesterUK
| | - Ari Waisman
- Institute for Molecular MedicineMedical University of Johannes Gutenberg‐University of MainzMainzGermany
| | | | - Werner Müller
- Faculty of Life SciencesUniversity of ManchesterManchesterUK
| |
Collapse
|
45
|
Gomez-Puerto MC, Folkerts H, Wierenga ATJ, Schepers K, Schuringa JJ, Coffer PJ, Vellenga E. Autophagy Proteins ATG5 and ATG7 Are Essential for the Maintenance of Human CD34(+) Hematopoietic Stem-Progenitor Cells. Stem Cells 2016; 34:1651-63. [PMID: 26930546 DOI: 10.1002/stem.2347] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 01/08/2016] [Indexed: 01/07/2023]
Abstract
Autophagy is a highly regulated catabolic process that involves sequestration and lysosomal degradation of cytosolic components such as damaged organelles and misfolded proteins. While autophagy can be considered to be a general cellular housekeeping process, it has become clear that it may also play cell type-dependent functional roles. In this study, we analyzed the functional importance of autophagy in human hematopoietic stem/progenitor cells (HSPCs), and how this is regulated during differentiation. Western blot-based analysis of LC3-II and p62 levels, as well as flow cytometry-based autophagic vesicle quantification, demonstrated that umbilical cord blood-derived CD34(+) /CD38(-) immature hematopoietic progenitors show a higher autophagic flux than CD34(+) /CD38(+) progenitors and more differentiated myeloid and erythroid cells. This high autophagic flux was critical for maintaining stem and progenitor function since knockdown of autophagy genes ATG5 or ATG7 resulted in reduced HSPC frequencies in vitro as well as in vivo. The reduction in HSPCs was not due to impaired differentiation, but at least in part due to reduced cell cycle progression and increased apoptosis. This is accompanied by increased expression of p53, proapoptotic genes BAX and PUMA, and the cell cycle inhibitor p21, as well as increased levels of cleaved caspase-3 and reactive oxygen species. Taken together, our data demonstrate that autophagy is an important regulatory mechanism for human HSCs and their progeny, reducing cellular stress and promoting survival. Stem Cells 2016;34:1651-1663.
Collapse
Affiliation(s)
- Maria Catalina Gomez-Puerto
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.,Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hendrik Folkerts
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University Groningen, Groningen, The Netherlands
| | - Albertus T J Wierenga
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University Groningen, Groningen, The Netherlands
| | - Koen Schepers
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan Jacob Schuringa
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University Groningen, Groningen, The Netherlands
| | - Paul J Coffer
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.,Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Edo Vellenga
- Department of Experimental Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University Groningen, Groningen, The Netherlands
| |
Collapse
|