1
|
Fan J, Wang X, Wang Y, Song J, Chen M, Weng C, Wang L, Chi Z, Zhang W. Dietary glutamine supplementation improves both Th1 and Th17 responses via CARD11-mTORC1 pathway in murine model of atopic dermatitis. Int Immunopharmacol 2024; 143:113316. [PMID: 39368135 DOI: 10.1016/j.intimp.2024.113316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
Glutamine (GLN) is considered an immunomodulatory nutrient, while caspase recruitment domain 11 (CARD11) is a susceptibility locus for atopic dermatitis (AD). T-cell antigen receptor (TCR)-stimulated GLN uptake requires CARD11. However, the specific pathogenesis of AD via GLN uptake remains unclear. This study aimed to elucidate the association between dietary GLN supplementation and the CARD11 pathway in the pathogenesis of AD, focusing on T helper type 1 (Th1) and Th17 cell expression in AD. Herein, wild-type (WT) mice with house dust mite epidermal-sensitized skin exhibited increased expression of interferon-gamma (IFN-gamma) and interleukin (IL)-17, whereas CARD11 deficiency impaired Th1 and Th17 responses at the same site. CARD11 is a key mediator of Th1 and Th17 expression in AD. Additionally, we suppressed mammalian target of rapamycin complex 1 (mTORC1) signaling, downstream of CARD11, to underscore the critical role of CARD11 in mediating Th1 and Th17 expression in AD. Further, dietary supplementation of GLN to CARD11-/- mice restored Th1 and Th17 responses, whereas inflammatory expression was reduced in WT mice, and p-CARD11 expression and mTORC1 signaling activity were increased in JPM50.6 cells and CARD11-/- mice. Upon inhibiting the GLN transporter, alanine-serine-cysteine transporter carrier 2 (ASCT2), we observed that the Th1 and Th17 response in AD was reduced. Conclusively, ASCT2-mediated GLN uptake improves the expression of Th1 and Th17 cells via CARD11-mTORC1 signaling pathway in AD, suggesting the potential of glutamine supplementation for AD treatment.
Collapse
Affiliation(s)
- Junwen Fan
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xiaoming Wang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yufei Wang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Jingjing Song
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Mingxin Chen
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Cuiye Weng
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Lei Wang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zailong Chi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| | - Weixi Zhang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
2
|
Layzell BS, Barbarulo A, van Loo G, Beyaert R, Seddon B. NF-κB regulated expression of A20 controls IKK dependent repression of RIPK1 induced cell death in activated T cells. Cell Death Differ 2024:10.1038/s41418-024-01383-6. [PMID: 39327505 DOI: 10.1038/s41418-024-01383-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
IKK signalling is essential for survival of thymocytes by repressing RIPK1 induced cell death rather than its canonical function of activating NF-κB. The role of IKK signalling in activated T cells is unclear. To investigate this, we analysed activation of IKK2 deficient T cells. While TCR triggering was normal, proliferation and expansion was profoundly impaired. This was not due to defective cell cycle progression, rather dividing T cells became sensitised to TNF induced cell death, since inhibition of RIPK1 kinase activity rescued cell survival. Gene expression analysis of activated IKK2 deficient T cells revealed defective expression of Tnfaip3, that encodes A20, a negative regulator of NF-κB. To test whether A20 expression was required to protect IKK2 deficient T cells from cell death, we generated mice with T cells lacking both A20 and IKK2. Doing this resulted in near complete loss of peripheral T cells, in contrast to mice lacking one or other gene. Strikingly, this phenotype was completely reversed by inactivation of RIPK1 kinase activity in vivo. Together, our data show that IKK signalling in activated T cells protects against RIPK1 dependent death, both by direct phosphorylation of RIPK1 and through NF-κB mediated induction of A20, that we identify for the first time as a key modulator of RIPK1 activity in T cells.
Collapse
Affiliation(s)
- By Scott Layzell
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, The Pears Building, Hampstead, London, UK
| | - Alessandro Barbarulo
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, The Pears Building, Hampstead, London, UK
| | - Geert van Loo
- VIB-UGent Center for Inflammation Research, UGent Department for Biomedical Molecular Biology, Unit of Molecular Signal Transduction in Inflammation, Gent, Belgium
| | - Rudi Beyaert
- VIB-UGent Center for Inflammation Research, UGent Department for Biomedical Molecular Biology, Unit of Molecular Signal Transduction in Inflammation, Gent, Belgium
| | - Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, The Pears Building, Hampstead, London, UK.
| |
Collapse
|
3
|
Alsaidalani AA, García-Solís B, Bukhari E, Van Den Rym A, López-Collazo E, Sánchez-Ramón S, Corvillo F, López-Lera A, de Andrés A, Martínez-Barricarte R, Perez de Diego R. Inherited Human BCL10 Deficiencies. J Clin Immunol 2023; 44:13. [PMID: 38129623 PMCID: PMC10966939 DOI: 10.1007/s10875-023-01619-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/26/2023] [Indexed: 12/23/2023]
Abstract
Human BCL10 deficiency causes combined immunodeficiency with bone marrow transplantation as its only curative option. To date, there are four homozygous mutations described in the literature that were identified in four unrelated patients. Here, we describe a fifth patient with a novel mutation and summarize what we have learned about BCL10 deficiency. Due to the severity of the disease, accurate knowledge of its clinical and immunological characteristics is instrumental for early diagnosis and adequate clinical management of the patients.
Collapse
Affiliation(s)
- Ashwag A Alsaidalani
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, 22252, Jeddah, Saudi Arabia
| | - Blanca García-Solís
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz University Hospital, 28046, Madrid, Spain
- Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, 28046, Madrid, Spain
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| | - Esraa Bukhari
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, 22252, Jeddah, Saudi Arabia
| | - Ana Van Den Rym
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz University Hospital, 28046, Madrid, Spain
- Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, 28046, Madrid, Spain
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain
| | - Eduardo López-Collazo
- Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, 28046, Madrid, Spain
| | - Silvia Sánchez-Ramón
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain
- Clinical Immunology Department and IdSSC, San Carlos Clinical Hospital, 28040, Madrid, Spain
| | - Fernando Corvillo
- IdiPAZ Institute for Health Research, La Paz University Hospital, CIBERER U-754, 28046, Madrid, Spain
| | - Alberto López-Lera
- IdiPAZ Institute for Health Research, La Paz University Hospital, CIBERER U-754, 28046, Madrid, Spain
| | - Ana de Andrés
- Immunology Department, Hospital Ramon y Cajal, 28034, Madrid, Spain
| | - Rubén Martínez-Barricarte
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Immunology, and Inflammation, Vanderbilt Institute for Infection, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Rebeca Perez de Diego
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, La Paz University Hospital, 28046, Madrid, Spain.
- Innate Immunity Group, IdiPAZ Institute for Health Research, La Paz University Hospital, 28046, Madrid, Spain.
- Interdepartmental Group of Immunodeficiencies, Madrid, Spain.
| |
Collapse
|
4
|
Boutboul D, Picard C, Latour S. Inborn errors of immunity underlying defective T-cell memory. Curr Opin Allergy Clin Immunol 2023; 23:491-499. [PMID: 37797193 DOI: 10.1097/aci.0000000000000946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
PURPOSE OF REVIEW T-cell memory is a complex process not well understood involving specific steps, pathways and different T-cell subpopulations. Inborn errors of immunity (IEIs) represent unique models to decipher some of these requirements in humans. More than 500 different IEIs have been reported to date, and recently a subgroup of monogenic disorders characterized by memory T-cell defects has emerged, providing novel insights into the pathways of T-cell memory generation and maintenance, although this new knowledge is mostly restricted to peripheral blood T-cell memory populations. RECENT FINDINGS This review draws up an inventory of the main and recent IEIs associated with T-cell memory defects and their mice models, with a particular focus on the nuclear factor kappa B (NF-κB) signalling pathway, including the scaffold protein capping protein regulator and myosin 1 linker 2 (CARMIL2) and the T-cell co-stimulatory molecules CD28 and OX-40. Besides NF-κB, IKZF1 (IKAROS), a key transcription factor of haematopoiesis and STAT3-dependent interleukin-6 signals involving the transcription factor ZNF341 also appear to be important for the generation of T cell memory. Somatic reversion mosaicism in memory T cells is documented for several gene defects supporting the critical role of these factors in the development of memory T cells with a potential clinical benefit. SUMMARY Systematic examination of T-cell memory subsets could be helpful in the diagnosis of IEIs.
Collapse
Affiliation(s)
- David Boutboul
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute
- Haematology department, Hospital Cochin, Assistance Publique-Hôpitaux de Paris (APHP)
- Université de Paris Cité
| | - Capucine Picard
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute
- Study Center for Primary Immunodeficiencies, Necker-Enfants Malades Hospital
- Université de Paris Cité
- Centre de références des déficits immunitaires Héréditaires (CEREDIH), Necker-Enfants Malades Hospital APHP, Paris, France
| | - Sylvain Latour
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute
- Université de Paris Cité
| |
Collapse
|
5
|
Carty F, Layzell S, Barbarulo A, Islam F, Webb LV, Seddon B. IKK promotes naïve T cell survival by repressing RIPK1-dependent apoptosis and activating NF-κB. Sci Signal 2023; 16:eabo4094. [PMID: 37368952 DOI: 10.1126/scisignal.abo4094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 06/07/2023] [Indexed: 06/29/2023]
Abstract
The inhibitor of κB kinase (IKK) complex regulates the activation of the nuclear factor κB (NF-κB) family of transcription factors. In addition, IKK represses extrinsic cell death pathways dependent on receptor-interacting serine/threonine-protein kinase 1 (RIPK1) by directly phosphorylating this kinase. Here, we showed that peripheral naïve T cells in mice required the continued expression of IKK1 and IKK2 for their survival; however, the loss of these cells was only partially prevented when extrinsic cell death pathways were blocked by either deleting Casp8 (which encodes the apoptosis-inducing caspase 8) or inhibiting the kinase activity of RIPK1. Inducible deletion of Rela (which encodes the NF-κB p65 subunit) in mature CD4+ T cells also resulted in loss of naïve CD4+ T cells and in reduced abundance of the interleukin-7 receptor (IL-7R) encoded by the NF-κB target Il7r, revealing an additional reliance upon NF-κB for the long-term survival of mature T cells. Together, these data indicate that the IKK-dependent survival of naïve CD4+ T cells depends on both repression of extrinsic cell death pathways and activation of an NF-κB-dependent survival program.
Collapse
Affiliation(s)
- Fiona Carty
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London NW3 2PP, UK
| | - Scott Layzell
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London NW3 2PP, UK
| | - Alessandro Barbarulo
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London NW3 2PP, UK
| | - Farjana Islam
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London NW3 2PP, UK
| | - Louise V Webb
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London NW3 2PP, UK
| | - Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London NW3 2PP, UK
| |
Collapse
|
6
|
Baronio M, Gazzurelli L, Rezzola S, Rossi S, Tessarin G, Marinoni M, Salpietro A, Fiore M, Moratto D, Chiarini M, Badolato R, Parolini S, Tabellini G, Lougaris V. CARD11 dominant negative mutation leads to altered human Natural Killer cell homeostasis. Immunobiology 2023; 228:152381. [PMID: 37086690 DOI: 10.1016/j.imbio.2023.152381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/10/2023] [Accepted: 03/28/2023] [Indexed: 04/07/2023]
Abstract
Dominant negative mutations in CARD11 have been reported in patients with immune dysregulation, severe atopic features, and variable T cell alterations. Data on Natural killer (NK) cells from affected patients are lacking. We report on a 12-year-old boy with severe atopic dermatitis, food induced anaphylaxis and hypogammaglobulinemia harbouring a novel de novo heterozygous variant c.169G > A; p.Glu57Lys in CARD11. The dominant negative effect of this mutation was confirmed on both CD4+ and CD8+. CTLA4+Foxp3+CD4+ Tregs were severely reduced. Patient's NK cells showed reduced expression of NKp46, NKG2D and CD69. Patient's CD56bright NK cells showed in vitro impaired production of IFN-γ. Steady state pS6 levels on patient's NK cells were increased and remained elevated upon IL2 + IL12 + IL18 overnight stimulation. Overall, the effect of CARD11 mutation on mTORC1 differs between T and NK cells. These findings may explain the increased susceptibility to viral infections and the reduced immune surveillance in affected patients.
Collapse
Affiliation(s)
- Manuela Baronio
- Pediatrics Clinic and "A. Nocivelli" Institute for Molecular Medicine, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Luisa Gazzurelli
- Pediatrics Clinic and "A. Nocivelli" Institute for Molecular Medicine, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Stefano Rossi
- Pediatrics Clinic and "A. Nocivelli" Institute for Molecular Medicine, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Giulio Tessarin
- Pediatrics Clinic and "A. Nocivelli" Institute for Molecular Medicine, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Maddalena Marinoni
- Paediatric Department, ASST-Sette Laghi, "F. Del Ponte" Hospital, Varese, Italy
| | - Annamaria Salpietro
- Pediatrics Clinic and "A. Nocivelli" Institute for Molecular Medicine, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Michele Fiore
- Primary Care Pediatrician, Local unit ASL3 "genovese", Genova, Italy
| | - Daniele Moratto
- Flow Cytometry Laboratory, Diagnostic Department, ASST Spedali Civili, Brescia, Italy
| | - Marco Chiarini
- Flow Cytometry Laboratory, Diagnostic Department, ASST Spedali Civili, Brescia, Italy
| | - Raffaele Badolato
- Pediatrics Clinic and "A. Nocivelli" Institute for Molecular Medicine, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Silvia Parolini
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Giovanna Tabellini
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Vassilios Lougaris
- Pediatrics Clinic and "A. Nocivelli" Institute for Molecular Medicine, Department of Clinical and Experimental Sciences, University of Brescia, ASST Spedali Civili of Brescia, Brescia, Italy.
| |
Collapse
|
7
|
Zheng Y, Yu M, Chen Y, Xue L, Zhu W, Fu G, Morris SW, Wen R, Wang D. CARD19, a Novel Regulator of the TAK1/NF-κB Pathway in Self-Reactive B Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1222-1235. [PMID: 36961449 PMCID: PMC10156913 DOI: 10.4049/jimmunol.2200639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 02/22/2023] [Indexed: 03/25/2023]
Abstract
The caspase recruitment domain family member (CARD)11-Bcl10-Malt1 signalosome controls TGF-β-activated kinase 1 (TAK1) activation and regulates BCR-induced NF-κB activation. In this study, we discovered that CARD19 interacted with TAK1 and inhibited TAB2-mediated TAK1 ubiquitination and activation. Although CARD19 deficiency in mice did not affect B cell development, it enhanced clonal deletion, receptor editing, and anergy of self-reactive B cells, and it reduced autoantibody production. Mechanistically, CARD19 deficiency increased BCR/TAK1-mediated NF-κB activation, leading to increased expression of transcription factors Egr2/3, as well as the E3 ubiquitin ligases c-Cbl/Cbl-b, which are known inducers of B cell tolerance in self-reactive B cells. RNA sequencing analysis revealed that although CARD19 deficiency did not affect the overall Ag-induced gene expression in naive B cells, it suppressed BCR signaling and increased hyporesponsiveness of self-reactive B cells. As a result, CARD19 deficiency prevented Bm12-induced experimental systemic lupus erythematosus. In summary, CARD19 negatively regulates BCR/TAK1-induced NF-κB activation and its deficiency increases Egr2/3 and c-Cbl/Cbl-b expression in self-reactive B cells, thereby enhancing B cell tolerance.
Collapse
Affiliation(s)
| | - Mei Yu
- Versiti Blood Research Institute, Milwaukee, WI
| | - Yuhong Chen
- Versiti Blood Research Institute, Milwaukee, WI
| | | | - Wen Zhu
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Guoping Fu
- Versiti Blood Research Institute, Milwaukee, WI
| | | | - Renren Wen
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Demin Wang
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
8
|
Sun L, Yang B, Peng Z, Yang T, Qin B, Ao J, Yang Y, Wang J, Zheng L, Xie H. Transcriptomics and Phenotypic Analysis of gpr56 Knockout in Zebrafish. Int J Mol Sci 2023; 24:ijms24097740. [PMID: 37175447 PMCID: PMC10178538 DOI: 10.3390/ijms24097740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
The adhesion G-protein-coupled receptor is a seven-transmembrane receptor protein with a complex structure. Impaired GPR56 has been found to cause developmental damage to the human brain, resulting in intellectual disability and motor dysfunction. To date, studies on gpr56 deficiency in zebrafish have been limited to the nervous system, and there have been no reports of its systemic effects on juvenile fish at developmental stages. In order to explore the function of gpr56 in zebrafish, the CRISPR/Cas9 gene-editing system was used to construct a gpr56-knockout zebrafish. Subsequently, the differentially expressed genes (DEGs) at the transcriptional level between the 3 days post fertilization (dpf) homozygotes of the gpr56 mutation and the wildtype zebrafish were analyzed via RNA-seq. The results of the clustering analysis, quantitative PCR (qPCR), and in situ hybridization demonstrated that the expression of innate immunity-related genes in the mutant was disordered, and multiple genes encoding digestive enzymes of the pancreatic exocrine glands were significantly downregulated in the mutant. Motor ability tests demonstrated that the gpr56-/- zebrafish were more active, and this change was more pronounced in the presence of cold and additional stimuli. In conclusion, our results revealed the effect of gpr56 deletion on the gene expression of juvenile zebrafish and found that the gpr56 mutant was extremely active, providing an important clue for studying the mechanism of gpr56 in the development of juvenile zebrafish.
Collapse
Affiliation(s)
- Luning Sun
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha 410081, China
| | - Boyu Yang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha 410081, China
| | - Zheng Peng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha 410081, China
| | - Tianle Yang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha 410081, China
| | - Bin Qin
- Heart Development Center, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Jieyu Ao
- Heart Development Center, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Yanqun Yang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha 410081, China
| | - Jingling Wang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha 410081, China
| | - Lan Zheng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha 410081, China
| | - Huaping Xie
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Changsha 410081, China
| |
Collapse
|
9
|
Xia X, Qu K, Wang Y, Sinding MHS, Wang F, Hanif Q, Ahmed Z, Lenstra JA, Han J, Lei C, Chen N. Global dispersal and adaptive evolution of domestic cattle: a genomic perspective. STRESS BIOLOGY 2023; 3:8. [PMID: 37676580 PMCID: PMC10441868 DOI: 10.1007/s44154-023-00085-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 03/26/2023] [Indexed: 09/08/2023]
Abstract
Domestic cattle have spread across the globe and inhabit variable and unpredictable environments. They have been exposed to a plethora of selective pressures and have adapted to a variety of local ecological and management conditions, including UV exposure, diseases, and stall-feeding systems. These selective pressures have resulted in unique and important phenotypic and genetic differences among modern cattle breeds/populations. Ongoing efforts to sequence the genomes of local and commercial cattle breeds/populations, along with the growing availability of ancient bovid DNA data, have significantly advanced our understanding of the genomic architecture, recent evolution of complex traits, common diseases, and local adaptation in cattle. Here, we review the origin and spread of domestic cattle and illustrate the environmental adaptations of local cattle breeds/populations.
Collapse
Affiliation(s)
- Xiaoting Xia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Kaixing Qu
- Academy of Science and Technology, Chuxiong Normal University, Chuxiong, 675000, China
| | - Yan Wang
- Qingdao Municipal Bureau of Agriculture and Rural Affairs, Qingdao, 266000, China
| | - Mikkel-Holger S Sinding
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, 1350, Denmark
| | - Fuwen Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Quratulain Hanif
- National Institute for Biotechnology and Genetic Engineering, Faisalabad, Pakistan
| | - Zulfiqar Ahmed
- Faculty of Veterinary and Animal Sciences, University of Poonch Rawalakot, Azad Jammu and Kashmir, 12350, Pakistan
| | - Johannes A Lenstra
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jianlin Han
- Livestock Genetic Program, International Livestock Research Institute (ILRI), Nairobi, 00100, Kenya
- CAAS-ILRI Joint Laboratory On Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| | - Ningbo Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
10
|
Masle-Farquhar E, Jeelall Y, White J, Bier J, Deenick EK, Brink R, Horikawa K, Goodnow CC. CARD11 gain-of-function mutation drives cell-autonomous accumulation of PD-1 + ICOS high activated T cells, T-follicular, T-regulatory and T-follicular regulatory cells. Front Immunol 2023; 14:1095257. [PMID: 36960072 PMCID: PMC10028194 DOI: 10.3389/fimmu.2023.1095257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Introduction Germline CARD11 gain-of-function (GOF) mutations cause B cell Expansion with NF-κB and T cell Anergy (BENTA) disease, whilst somatic GOF CARD11 mutations recur in diffuse large B cell lymphoma (DLBCL) and in up to 30% of the peripheral T cell lymphomas (PTCL) adult T cell leukemia/lymphoma (ATL), cutaneous T cell lymphoma (CTCL) and Sezary Syndrome. Despite their frequent acquisition by PTCL, the T cell-intrinsic effects of CARD11 GOF mutations are poorly understood. Methods Here, we studied B and T lymphocytes in mice with a germline Nethyl-N-nitrosourea (ENU)-induced Card11M365K mutation identical to a mutation identified in DLBCL and modifying a conserved region of the CARD11 coiled-coil domain recurrently mutated in DLBCL and PTCL. Results and discussion Our results demonstrate that CARD11.M365K is a GOF protein that increases B and T lymphocyte activation and proliferation following antigen receptor stimulation. Germline Card11M365K mutation was insufficient alone to cause B or T-lymphoma, but increased accumulation of germinal center (GC) B cells in unimmunized and immunized mice. Card11M365K mutation caused cell-intrinsic over-accumulation of activated T cells, T regulatory (TREG), T follicular (TFH) and T follicular regulatory (TFR) cells expressing increased levels of ICOS, CTLA-4 and PD-1 checkpoint molecules. Our results reveal CARD11 as an important, cell-autonomous positive regulator of TFH, TREG and TFR cells. They highlight T cell-intrinsic effects of a GOF mutation in the CARD11 gene, which is recurrently mutated in T cell malignancies that are often aggressive and associated with variable clinical outcomes.
Collapse
Affiliation(s)
- Etienne Masle-Farquhar
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Clinical, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- *Correspondence: Etienne Masle-Farquhar, ; Yogesh Jeelall,
| | - Yogesh Jeelall
- John Curtin School of Medical Research, Immunology Department, The Australian National University, Canberra, ACT, Australia
- *Correspondence: Etienne Masle-Farquhar, ; Yogesh Jeelall,
| | - Jacqueline White
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Clinical, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Julia Bier
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Clinical, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Elissa K. Deenick
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Clinical, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Robert Brink
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Clinical, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Keisuke Horikawa
- John Curtin School of Medical Research, Immunology Department, The Australian National University, Canberra, ACT, Australia
| | - Christopher Carl Goodnow
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- Cellular Genomics Futures Institute, University of New South Wales, Sydney, Australia
| |
Collapse
|
11
|
Gui Z, Zhang Y, Zhang A, Xia W, Jia Z. CARMA3: A potential therapeutic target in non-cancer diseases. Front Immunol 2022; 13:1057980. [PMID: 36618379 PMCID: PMC9815110 DOI: 10.3389/fimmu.2022.1057980] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Caspase recruitment domain and membrane-associated guanylate kinase-like protein 3 (CARMA3) is a scaffold protein widely expressed in non-hematopoietic cells. It is encoded by the caspase recruitment domain protein 10 (CARD10) gene. CARMA3 can form a CARMA3-BCL10-MALT1 complex by recruiting B cell lymphoma 10 (BCL10) and mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1), thereby activating nuclear factor-κB (NF-κB), a key transcription factor that involves in various biological responses. CARMA3 mediates different receptors-dependent signaling pathways, including G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs). Inappropriate expression and activation of GPCRs and/or RTKs/CARMA3 signaling lead to the pathogenesis of human diseases. Emerging studies have reported that CARMA3 mediates the development of various types of cancers. Moreover, CARMA3 and its partners participate in human non-cancer diseases, including atherogenesis, abdominal aortic aneurysm, asthma, pulmonary fibrosis, liver fibrosis, insulin resistance, inflammatory bowel disease, and psoriasis. Here we provide a review on its structure, regulation, and molecular function, and further highlight recent findings in human non-cancerous diseases, which will provide a novel therapeutic target.
Collapse
Affiliation(s)
- Zhen Gui
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Zhang
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Weiwei Xia
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China,Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China,*Correspondence: Zhanjun Jia, ; Weiwei Xia,
| | - Zhanjun Jia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China,*Correspondence: Zhanjun Jia, ; Weiwei Xia,
| |
Collapse
|
12
|
Pomerantz JL, Milner JD, Snow AL. Elevated IgE from attenuated CARD11 signaling: lessons from atopic mice and humans. Curr Opin Immunol 2022; 79:102255. [PMID: 36334349 PMCID: PMC10424059 DOI: 10.1016/j.coi.2022.102255] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/17/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022]
Abstract
CARD11 encodes a large scaffold protein responsible for integrating antigen-receptor engagement with downstream signaling to NF-kB and other outputs in lymphocytes. Over the past 10 years, several human-inborn errors of immunity have been linked to pathogenic CARD11 mutations. Most recently, severe atopic patients were discovered that carried heterozygous dominant-negative CARD11 mutations. Here, we review the mechanistic connections between attenuated CARD11 signaling, elevated IgE, and atopy, comparing and contrasting key insights from both human patients and murine models. Continued investigation of abnormal CARD11 signaling in both contexts should inform novel therapeutic strategies to combat allergic pathogenesis.
Collapse
Affiliation(s)
- Joel L Pomerantz
- Department of Biological Chemistry, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joshua D Milner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrew L Snow
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
13
|
Urdinez L, Erra L, Palma AM, Mercogliano MF, Fernandez JB, Prieto E, Goris V, Bernasconi A, Sanz M, Villa M, Bouso C, Caputi L, Quesada B, Solis D, Aguirre Bruzzo A, Katsicas MM, Galluzzo L, Weyersberg C, Bocian M, Bujan MM, Oleastro M, Almejun MB, Danielian S. Expanding spectrum, intrafamilial diversity, and therapeutic challenges from 15 patients with heterozygous CARD11-associated diseases: A single center experience. Front Immunol 2022; 13:1020927. [PMID: 36405754 PMCID: PMC9668901 DOI: 10.3389/fimmu.2022.1020927] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2023] Open
Abstract
CARD11-associated diseases are monogenic inborn errors of immunity involving immunodeficiency, predisposition to malignancy and immune dysregulation such as lymphoproliferation, inflammation, atopic and autoimmune manifestations. Defects in CARD11 can present as mutations that confer a complete or a partial loss of function (LOF) or contrarily, a gain of function (GOF) of the affected gene product. We report clinical characteristics, immunophenotypes and genotypes of 15 patients from our center presenting with CARD11-associated diseases. Index cases are pediatric patients followed in our immunology division who had access to next generation sequencing studies. Variant significance was defined by functional analysis in cultured cells transfected with a wild type and/or with mutated hCARD11 constructs. Cytoplasmic aggregation of CARD11 products was evaluated by immunofluorescence. Nine index patients with 9 unique heterozygous CARD11 variants were identified. At the time of the identification, 7 variants previously unreported required functional validation. Altogether, four variants showed a GOF effect as well a spontaneous aggregation in the cytoplasm, leading to B cell expansion with NF-κB and T cell anergy (BENTA) diagnosis. Additional four variants showing a LOF activity were considered as causative of CARD11-associated atopy with dominant interference of NF-kB signaling (CADINS). The remaining variant exhibited a neutral functional assay excluding its carrier from further analysis. Family segregation studies expanded to 15 individuals the number of patients presenting CARD11-associated disease. A thorough clinical, immunophenotypical, and therapeutic management evaluation was performed on these patients (5 BENTA and 10 CADINS). A remarkable variability of disease expression was clearly noted among BENTA as well as in CADINS patients, even within multiplex families. Identification of novel CARD11 variants required functional studies to validate their pathogenic activity. In our cohort BENTA phenotype exhibited a more severe and expanded clinical spectrum than previously reported, e.g., severe hematological and extra hematological autoimmunity and 3 fatal outcomes. The growing number of patients with dysmorphic facial features strengthen the inclusion of extra-immune characteristics as part of the CADINS spectrum. CARD11-associated diseases represent a challenging group of disorders from the diagnostic and therapeutic standpoint, especially BENTA cases that can undergo a more severe progression than previously described.
Collapse
Affiliation(s)
- Luciano Urdinez
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Lorenzo Erra
- Laboratorio de Biofisicoquímica de Proteínas, Departamento de Química Biológica, Instituto de Quimica Biologica de Facultad de Ciencias Biologicas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Laboratorio de Genética en Endocrinología, Instituto de Biociencias, Biotecnologia y Biologia Translacional (IB3), Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alejandro M. Palma
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - María F. Mercogliano
- Laboratorio de Biofisicoquímica de Proteínas, Departamento de Química Biológica, Instituto de Quimica Biologica de Facultad de Ciencias Biologicas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Laboratorio de Genética en Endocrinología, Instituto de Biociencias, Biotecnologia y Biologia Translacional (IB3), Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Julieta Belén Fernandez
- Laboratorio de Biofisicoquímica de Proteínas, Departamento de Química Biológica, Instituto de Quimica Biologica de Facultad de Ciencias Biologicas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Laboratorio de Genética en Endocrinología, Instituto de Biociencias, Biotecnologia y Biologia Translacional (IB3), Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Emma Prieto
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Verónica Goris
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Andrea Bernasconi
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Marianela Sanz
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Mariana Villa
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Carolina Bouso
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Lucia Caputi
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Belen Quesada
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Daniel Solis
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Anabel Aguirre Bruzzo
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Maria Martha Katsicas
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Laura Galluzzo
- Servicio de Anatomía Patológica, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Christian Weyersberg
- Servicio de Gastroenterología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Marcela Bocian
- Servicio de Dermatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Maria Marta Bujan
- Servicio de Dermatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - Matías Oleastro
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | - María B. Almejun
- Laboratorio de Biofisicoquímica de Proteínas, Departamento de Química Biológica, Instituto de Quimica Biologica de Facultad de Ciencias Biologicas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Laboratorio de Genética en Endocrinología, Instituto de Biociencias, Biotecnologia y Biologia Translacional (IB3), Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Silvia Danielian
- Servicio de Inmunología y Reumatología, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| |
Collapse
|
14
|
Kanduc D. SARS-CoV-2-Induced Immunosuppression: A Molecular Mimicry Syndrome. Glob Med Genet 2022; 9:191-199. [PMID: 35846107 PMCID: PMC9282940 DOI: 10.1055/s-0042-1748170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background
Contrary to immunological expectations, decay of adaptive responses against severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) characterizes recovered patients compared with patients who had a severe disease course or died following SARS-CoV-2 infection. This raises the question of the causes of the virus-induced immune immunosuppression. Searching for molecular link(s) between SARS-CoV-2 immunization and the decay of the adaptive immune responses, SARS-CoV-2 proteome was analyzed for molecular mimicry with human proteins related to immunodeficiency. The aim was to verify the possibility of cross-reactions capable of destroying the adaptive immune response triggered by SARS-CoV-2.
Materials and Methods
Human immunodeficiency–related proteins were collected from UniProt database and analyzed for sharing of minimal immune determinants with the SARS-CoV-2 proteome.
Results
Molecular mimicry and consequent potential cross-reactivity exist between SARS-CoV-2 proteome and human immunoregulatory proteins such as nuclear factor kappa B (NFKB), and variable diversity joining V(D)J recombination-activating gene (RAG).
Conclusion
The data (1) support molecular mimicry and the associated potential cross-reactivity as a mechanism that can underlie self-reactivity against proteins involved in B- and T-cells activation/development, and (2) suggest that the extent of the immunosuppression is dictated by the extent of the immune responses themselves. The higher the titer of the immune responses triggered by SARS-CoV-2 immunization, the more severe can be the cross-reactions against the human immunodeficiency–related proteins, the more severe the immunosuppression. Hence, SARS-CoV-2-induced immunosuppression can be defined as a molecular mimicry syndrome. Clinically, the data imply that booster doses of SARS-CoV-2 vaccines may have opposite results to those expected.
Collapse
Affiliation(s)
- Darja Kanduc
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| |
Collapse
|
15
|
Vaseghi-Shanjani M, Snow AL, Margolis DJ, Latrous M, Milner JD, Turvey SE, Biggs CM. Atopy as Immune Dysregulation: Offender Genes and Targets. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:1737-1756. [PMID: 35680527 DOI: 10.1016/j.jaip.2022.04.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 06/15/2023]
Abstract
Allergic diseases are a heterogeneous group of disorders resulting from exaggerated type 2 inflammation. Although typically viewed as polygenic multifactorial disorders caused by the interaction of several genes with the environment, we have come to appreciate that allergic diseases can also be caused by monogenic variants affecting the immune system and the skin epithelial barrier. Through a myriad of genetic association studies and high-throughput sequencing tools, many monogenic and polygenic culprits of allergic diseases have been described. Identifying the genetic causes of atopy has shaped our understanding of how these conditions occur and how they may be treated and even prevented. Precision diagnostic tools and therapies that address the specific molecular pathways implicated in allergic inflammation provide exciting opportunities to improve our care for patients across the field of allergy and immunology. Here, we highlight offender genes implicated in polygenic and monogenic allergic diseases and list targeted therapeutic approaches that address these disrupted pathways.
Collapse
Affiliation(s)
- Maryam Vaseghi-Shanjani
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; Experimental Medicine Program, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Md
| | - David J Margolis
- Department of Dermatology and Dermatologic Surgery, University of Pennsylvania Medical Center, Philadelphia, Pa; Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Medical Center, Philadelphia, Pa
| | - Meriem Latrous
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joshua D Milner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Stuart E Turvey
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; Experimental Medicine Program, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Catherine M Biggs
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; St Paul's Hospital, Vancouver, British Columbia, Canada.
| |
Collapse
|
16
|
Cunha C, Koike T, Seki Y, Yamamoto M, Iwashima M. Schnurri 3 promotes Th2 cytokine production during the late phase of T-cell antigen stimulation. Eur J Immunol 2022; 52:1077-1094. [PMID: 35490426 PMCID: PMC9276650 DOI: 10.1002/eji.202149633] [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: 09/15/2021] [Revised: 03/22/2022] [Accepted: 04/28/2022] [Indexed: 11/29/2022]
Abstract
Th1 and Th2 polarization is determined by the coordination of numerous factors including the affinity and strength of the antigen-receptor interaction, predominant cytokine environment, and costimulatory molecules present. Here, we show that Schnurri (SHN) proteins have distinct roles in Th1 and Th2 polarization. SHN2 was previously found to block the induction of GATA3 and Th2 differentiation. We found that, in contrast to SHN2, SHN3 is critical for IL-4 production and Th2 polarization. Strength of stimulation controls SHN2 and SHN3 expression patterns, where higher doses of antigen receptor stimulation promoted SHN3 expression and IL-4 production, along with repression of SHN2 expression. SHN3-deficient T cells showed a substantial defect in IL-4 production and expression of AP-1 components, particularly c-Jun and Jun B. This loss of early IL-4 production led to reduced GATA3 expression and impaired Th2 differentiation. Together, these findings uncover SHN3 as a novel, critical regulator of Th2 development.
Collapse
Affiliation(s)
- Christina Cunha
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
| | - Toru Koike
- Department of Biology, Faculty of ScienceShizuoka UniversityShizuokaJapan
| | - Yoichi Seki
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
- Van Kampen Cardiovascular Research Laboratory, Department of Thoracic and Cardiovascular Surgery, Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| | - Mutsumi Yamamoto
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
- Van Kampen Cardiovascular Research Laboratory, Department of Thoracic and Cardiovascular Surgery, Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| | - Makio Iwashima
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
- Van Kampen Cardiovascular Research Laboratory, Department of Thoracic and Cardiovascular Surgery, Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| |
Collapse
|
17
|
Bedsaul JR, Shah N, Hutcherson SM, Pomerantz JL. Mechanistic impact of oligomer poisoning by dominant-negative CARD11 variants. iScience 2022; 25:103810. [PMID: 35198875 PMCID: PMC8844825 DOI: 10.1016/j.isci.2022.103810] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/10/2021] [Accepted: 01/19/2022] [Indexed: 11/25/2022] Open
Abstract
The CARD11 scaffold controls antigen receptor signaling to NF-κB, JNK, and mTOR. Three classes of germline mutations in CARD11 cause Primary Immunodeficiency, including homozygous loss-of-function (LOF) mutations in CARD11 deficiency, heterozygous gain-of-function (GOF) mutations in BENTA disease, and heterozygous dominant-negative LOF mutations in CADINS. Here, we characterize LOF CARD11 mutants with a range of dominant-negative activities to identify the mechanistic properties that cause these variants to exert dominant effects when heterozygous. We find that strong dominant negatives can poison signaling from mixed wild-type:mutant oligomers at two steps in the CARD11 signaling cycle, at the Opening Step and at the Cofactor Association Step. Our findings provide evidence that CARD11 oligomer subunits cooperate in at least two steps during antigen receptor signaling and reveal how different LOF mutations in the same oligomeric signaling hub may cause disease with different inheritance patterns.
Collapse
Affiliation(s)
- Jacquelyn R. Bedsaul
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Neha Shah
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shelby M. Hutcherson
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joel L. Pomerantz
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
18
|
Shen Y, Boulton APR, Yellon RL, Cook MC. Skin manifestations of inborn errors of NF-κB. Front Pediatr 2022; 10:1098426. [PMID: 36733767 PMCID: PMC9888762 DOI: 10.3389/fped.2022.1098426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 12/23/2022] [Indexed: 01/18/2023] Open
Abstract
More than 400 single gene defects have been identified as inborn errors of immunity, including many arising from genes encoding proteins that affect NF-κB activity. We summarise the skin phenotypes in this subset of disorders and provide an overview of pathogenic mechanisms. NF-κB acts cell-intrinsically in basal epithelial cells during differentiation of skin appendages, influences keratinocyte proliferation and survival, and both responses to and amplification of inflammation, particularly TNF. Skin phenotypes include ectodermal dysplasia, reduction and hyperproliferation of keratinocytes, and aberrant recruitment of inflammatory cells, which often occur in combination. Phenotypes conferred by these rare monogenic syndromes often resemble those observed with more common defects. This includes oral and perineal ulceration and pustular skin disease as occurs with Behcet's disease, hyperkeratosis with microabscess formation similar to psoriasis, and atopic dermatitis. Thus, these genotype-phenotype relations provide diagnostic clues for this subset of IEIs, and also provide insights into mechanisms of more common forms of skin disease.
Collapse
Affiliation(s)
- Yitong Shen
- Department of Immunology, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Anne P R Boulton
- Department of Immunology, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Robert L Yellon
- Department of Immunology, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Matthew C Cook
- Department of Immunology, Cambridge University Hospitals, Cambridge, United Kingdom.,Centre for Personalised Immunology, Australian National University, Canberra, Australia.,Cambridge Institute of Therapeutic Immunology and Infectious Disease, and Department of Medicine, University of Cambridge, United Kingdom
| |
Collapse
|
19
|
Zhao P, Meng Q, Huang Y, Zhang L, Luo S, Zhang X, Tan L, Zhou A, Xiong H, He X. Identification and Characterization of a Germline Mutation in CARD11 From a Chinese Case of B Cell Expansion With NF-κB and T Cell Anergy. Front Immunol 2021; 12:676386. [PMID: 34557185 PMCID: PMC8453161 DOI: 10.3389/fimmu.2021.676386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/23/2021] [Indexed: 11/25/2022] Open
Abstract
B cell expansion with NF-κB and T cell anergy (BENTA) is a rare primary immunodeficiency disorder caused by gain-of-function (GOF) mutations in the CARD11 gene. Affected patients present with persistent B cell lymphocytosis in early childhood paired with lymphadenopathy and splenomegaly. Until now only six activating mutations from 14 patients have been reported in CARD11. Here we report a patient from China with polyclonal B cell lymphocytosis and frequent infections in early life. A heterozygous mutation (c.377G>A, G126D) in exon 5 of CARD11 gene (NM_032415) was identified by whole exome sequencing. In vitro functional studies showed that the G126D mutation is associated with increased expression of CARD11 and NF-κB activation in Hela cells. Flow cytometry analysis indicated NK cell activity and CD107a degranulation of the patient were decreased. RNA sequencing analysis showed that a number of genes in NF-κB pathway increased while those involved in NK cell activity and degranulation were down-regulated. In summary, our work identified a de novo germline GOF mutation in CARD11 with functional evidence of BENTA.
Collapse
Affiliation(s)
- Peiwei Zhao
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Qingjie Meng
- Department of Clinical Laboratory, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Yufeng Huang
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Lei Zhang
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Sukun Luo
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Xiankai Zhang
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Li Tan
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Aifen Zhou
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Hao Xiong
- Department of Hematology & Oncology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Xuelian He
- Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| |
Collapse
|
20
|
Hutcherson SM, Bedsaul JR, Pomerantz JL. Pathway-Specific Defects in T, B, and NK Cells and Age-Dependent Development of High IgE in Mice Heterozygous for a CADINS-Associated Dominant Negative CARD11 Allele. THE JOURNAL OF IMMUNOLOGY 2021; 207:1150-1164. [PMID: 34341167 DOI: 10.4049/jimmunol.2001233] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 06/19/2021] [Indexed: 12/13/2022]
Abstract
CARD11 is a multidomain scaffold protein required for normal activation of NF-κB, JNK, and mTOR during Ag receptor signaling. Germline CARD11 mutations cause at least three types of primary immunodeficiency including CARD11 deficiency, B cell expansion with NF-κB and T cell anergy (BENTA), and CARD11-associated atopy with dominant interference of NF-κB signaling (CADINS). CADINS is uniquely caused by heterozygous loss-of-function CARD11 alleles that act as dominant negatives. CADINS patients present with frequent respiratory and skin infections, asthma, allergies, and atopic dermatitis. However, precisely how a heterozygous dominant negative CARD11 allele leads to the development of this CADINS-specific cluster of symptoms remains poorly understood. To address this, we generated mice expressing the CARD11 R30W allele originally identified in patients. We find that CARD11R30W/+ mice exhibit impaired signaling downstream of CARD11 that leads to defects in T, B, and NK cell function and immunodeficiency. CARD11R30W/+ mice develop elevated serum IgE levels with 50% penetrance that becomes more pronounced with age, but do not develop spontaneous atopic dermatitis. CARD11R30W/+ mice display reduced regulatory T cell numbers, but not the Th2 expansion observed in other mice with diminished CARD11 activity. Interestingly, the presence of mixed CARD11 oligomers in CARD11R30W/+ mice causes more severe signaling defects in T cells than in B cells, and specifically impacts IFN-γ production by NK cells, but not NK cell cytotoxicity. Our findings help explain the high susceptibility of CADINS patients to infection and suggest that the development of high serum IgE is not sufficient to induce overt atopic symptoms.
Collapse
Affiliation(s)
- Shelby M Hutcherson
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jacquelyn R Bedsaul
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Joel L Pomerantz
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
21
|
Mechanistic understanding of the combined immunodeficiency in complete human CARD11 deficiency. J Allergy Clin Immunol 2021; 148:1559-1574.e13. [PMID: 33872653 DOI: 10.1016/j.jaci.2021.04.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 04/01/2021] [Accepted: 04/07/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Germline pathogenic variants impairing the caspase recruitment domain family member 11 (CARD11)-B cell chronic lymphocytic leukemia/lymphoma 10 (BCL10)-MALT1 paracaspase (MALT1) (CBM) complex are associated with diverse human diseases including combined immunodeficiency (CID), atopy, and lymphoproliferation. However, the impact of CARD11 deficiency on human B-cell development, signaling, and function is incompletely understood. OBJECTIVES This study sought to determine the cellular, immunological, and biochemical basis of disease for 2 unrelated patients who presented with profound CID associated with viral and fungal respiratory infections, interstitial lung disease, and severe colitis. METHODS Patients underwent next-generation sequencing, immunophenotyping by flow cytometry, signaling assays by immunoblot, and transcriptome profiling by RNA-sequencing. RESULTS Both patients carried identical novel pathogenic biallelic loss-of-function variants in CARD11 (c.2509C>T; p.Arg837∗) leading to undetectable protein expression. This variant prevented CBM complex formation, severely impairing the activation of nuclear factor-κB, c-Jun N-terminal kinase, and MALT1 paracaspase activity in B and T cells. This functional defect resulted in a developmental block in B cells at the naive and type 1 transitional B-cell stage and impaired circulating T follicular helper cell (cTFH) development, which was associated with impaired antibody responses and absent germinal center structures on lymph node histology. Transcriptomics indicated that CARD11-dependent signaling is essential for immune signaling pathways involved in the development of these cells. Both patients underwent hematopoietic stem cell transplantations, which led to functional normalization. CONCLUSIONS Complete human CARD11 deficiency causes profound CID by impairing naive/type 1 B-cell and cTFH cell development and abolishing activation of MALT1 paracaspase, NF-κB, and JNK activity. Hematopoietic stem cell transplantation functionally restores impaired signaling pathways.
Collapse
|
22
|
TIGIT/CD226 Axis Regulates Anti-Tumor Immunity. Pharmaceuticals (Basel) 2021; 14:ph14030200. [PMID: 33670993 PMCID: PMC7997242 DOI: 10.3390/ph14030200] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Tumors escape immune surveillance by inducing various immunosuppressive pathways, including the activation of inhibitory receptors on tumor-infiltrating T cells. While monoclonal antibodies (mAbs) blocking programmed cell death 1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) have been approved for multiple cancer indications, only a subset of patients benefit from immune checkpoint blockade therapies, highlighting the need for additional approaches. Therefore, the identification of new target molecules acting in distinct or complementary pathways in monotherapy or combination therapy with PD-1/PD-L1 blockade is gaining immense interest. T cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibitory motif (ITIM) domains (TIGIT) has received considerable attention in cancer immunotherapy. Recently, anti-TIGIT mAb (tiragolumab) has demonstrated promising clinical efficacy in non-small cell lung cancer treatment when combined with an anti-PD-L1 drug (Tecentriq), leading to phase III trial initiation. TIGIT is expressed mainly on T and natural killer cells; it functions as an inhibitory checkpoint receptor, thereby limiting adaptive and innate immunity. CD226 competes for binding with the same ligands with TIGIT but delivers a positive stimulatory signal to the immune cells. This review discusses the recent discoveries regarding the roles of TIGIT and CD226 in immune cell function and their potential application in cancer immunotherapy.
Collapse
|
23
|
Meitlis I, Allenspach EJ, Bauman BM, Phan IQ, Dabbah G, Schmitt EG, Camp ND, Torgerson TR, Nickerson DA, Bamshad MJ, Hagin D, Luthers CR, Stinson JR, Gray J, Lundgren I, Church JA, Butte MJ, Jordan MB, Aceves SS, Schwartz DM, Milner JD, Schuval S, Skoda-Smith S, Cooper MA, Starita LM, Rawlings DJ, Snow AL, James RG. Multiplexed Functional Assessment of Genetic Variants in CARD11. Am J Hum Genet 2020; 107:1029-1043. [PMID: 33202260 PMCID: PMC7820631 DOI: 10.1016/j.ajhg.2020.10.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/27/2020] [Indexed: 12/28/2022] Open
Abstract
Genetic testing has increased the number of variants identified in disease genes, but the diagnostic utility is limited by lack of understanding variant function. CARD11 encodes an adaptor protein that expresses dominant-negative and gain-of-function variants associated with distinct immunodeficiencies. Here, we used a "cloning-free" saturation genome editing approach in a diploid cell line to simultaneously score 2,542 variants for decreased or increased function in the region of CARD11 associated with immunodeficiency. We also described an exon-skipping mechanism for CARD11 dominant-negative activity. The classification of reported clinical variants was sensitive (94.6%) and specific (88.9%), which rendered the data immediately useful for interpretation of seven coding and splicing variants implicated in immunodeficiency found in our clinic. This approach is generalizable for variant interpretation in many other clinically actionable genes, in any relevant cell type.
Collapse
Affiliation(s)
- Iana Meitlis
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Eric J Allenspach
- Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - Bradly M Bauman
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Isabelle Q Phan
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Gina Dabbah
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Erica G Schmitt
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, MO 63130, USA
| | - Nathan D Camp
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | | | - Deborah A Nickerson
- Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - Michael J Bamshad
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - David Hagin
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, University of Tel Aviv, Tel Aviv 62919, Israel
| | - Christopher R Luthers
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Jeffrey R Stinson
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Jessica Gray
- Divisions of Immunobiology, and Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | | | - Joseph A Church
- Department of Pediatrics, Keck School of Medicine, University of Southern California and Children's Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Manish J Butte
- Division of Immunology, Allergy, and Rheumatology, Department of Pediatrics, University of California Los Angeles, Los Angeles, CA 90404, USA
| | - Mike B Jordan
- Divisions of Immunobiology, and Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Seema S Aceves
- Division of Allergy Immunology, Departments of Pediatrics and Medicine, University of California, San Diego, and Rady Children's Hospital, San Diego, CA 92123, USA
| | | | - Joshua D Milner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Susan Schuval
- Department of Pediatrics, Stonybrook University, Stony Brook, NY 11794, USA
| | - Suzanne Skoda-Smith
- Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Megan A Cooper
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, MO 63130, USA
| | - Lea M Starita
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - David J Rawlings
- Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Andrew L Snow
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Richard G James
- Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
24
|
Blanchett S, Boal-Carvalho I, Layzell S, Seddon B. NF-κB and Extrinsic Cell Death Pathways - Entwined Do-or-Die Decisions for T cells. Trends Immunol 2020; 42:76-88. [PMID: 33246882 DOI: 10.1016/j.it.2020.10.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/09/2020] [Accepted: 10/27/2020] [Indexed: 12/23/2022]
Abstract
NF-κB signaling is required at multiple stages of T cell development and function. The NF-κB pathway integrates signals from many receptors and involves diverse adapters and kinases. Recent advances demonstrate that kinases controlling NF-κB activation, such as the IKK complex, serve dual independent functions because they also control cell death checkpoints. Survival functions previously attributed to NF-κB are in fact mediated by these upstream kinases by novel mechanisms. This new understanding has led to a refined view of how NF-κB and cell death signaling are interlinked and how they regulate cell fate. We discuss how NF-κB activation and control of cell death signaling by common upstream triggers cooperate to regulate different aspects of T cell development and function.
Collapse
Affiliation(s)
- Sam Blanchett
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, UK
| | - Ines Boal-Carvalho
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, UK
| | - Scott Layzell
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, UK
| | - Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, UK.
| |
Collapse
|
25
|
The mosaic genome of indigenous African cattle as a unique genetic resource for African pastoralism. Nat Genet 2020; 52:1099-1110. [PMID: 32989325 DOI: 10.1038/s41588-020-0694-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 08/18/2020] [Indexed: 02/07/2023]
Abstract
Cattle pastoralism plays a central role in human livelihood in Africa. However, the genetic history of its success remains unknown. Here, through whole-genome sequence analysis of 172 indigenous African cattle from 16 breeds representative of the main cattle groups, we identify a major taurine × indicine cattle admixture event dated to circa 750-1,050 yr ago, which has shaped the genome of today's cattle in the Horn of Africa. We identify 16 loci linked to African environmental adaptations across crossbred animals showing an excess of taurine or indicine ancestry. These include immune-, heat-tolerance- and reproduction-related genes. Moreover, we identify one highly divergent locus in African taurine cattle, which is putatively linked to trypanotolerance and present in crossbred cattle living in trypanosomosis-infested areas. Our findings indicate that a combination of past taurine and recent indicine admixture-derived genetic resources is at the root of the present success of African pastoralism.
Collapse
|
26
|
Cheng J, Klei LR, Hubel NE, Zhang M, Schairer R, Maurer LM, Klei HB, Kang H, Concel VJ, Delekta PC, Dang EV, Mintz MA, Baens M, Cyster JG, Parameswaran N, Thome M, Lucas PC, McAllister-Lucas LM. GRK2 suppresses lymphomagenesis by inhibiting the MALT1 proto-oncoprotein. J Clin Invest 2020; 130:1036-1051. [PMID: 31961340 DOI: 10.1172/jci97040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/06/2019] [Indexed: 12/11/2022] Open
Abstract
Antigen receptor-dependent (AgR-dependent) stimulation of the NF-κB transcription factor in lymphocytes is a required event during adaptive immune response, but dysregulated activation of this signaling pathway can lead to lymphoma. AgR stimulation promotes assembly of the CARMA1-BCL10-MALT1 complex, wherein MALT1 acts as (a) a scaffold to recruit components of the canonical NF-κB machinery and (b) a protease to cleave and inactivate specific substrates, including negative regulators of NF-κB. In multiple lymphoma subtypes, malignant B cells hijack AgR signaling pathways to promote their own growth and survival, and inhibiting MALT1 reduces the viability and growth of these tumors. As such, MALT1 has emerged as a potential pharmaceutical target. Here, we identified G protein-coupled receptor kinase 2 (GRK2) as a new MALT1-interacting protein. We demonstrated that GRK2 binds the death domain of MALT1 and inhibits MALT1 scaffolding and proteolytic activities. We found that lower GRK2 levels in activated B cell-type diffuse large B cell lymphoma (ABC-DLBCL) are associated with reduced survival, and that GRK2 knockdown enhances ABC-DLBCL tumor growth in vitro and in vivo. Together, our findings suggest that GRK2 can function as a tumor suppressor by inhibiting MALT1 and provide a roadmap for developing new strategies to inhibit MALT1-dependent lymphomagenesis.
Collapse
Affiliation(s)
| | | | - Nathaniel E Hubel
- Department of Pediatrics and.,Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Ming Zhang
- Department of Biochemistry, Center of Immunity and Infection, University of Lausanne, Epalinges, Switzerland
| | - Rebekka Schairer
- Department of Biochemistry, Center of Immunity and Infection, University of Lausanne, Epalinges, Switzerland
| | | | | | - Heejae Kang
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | - Phillip C Delekta
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Eric V Dang
- Department of Biophysics and Biochemistry, UCSF, San Francisco, California, USA
| | - Michelle A Mintz
- Department of Biophysics and Biochemistry, UCSF, San Francisco, California, USA
| | - Mathijs Baens
- Human Genome Laboratory, VIB Center for the Biology of Disease, and.,Center for Human Genetics, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jason G Cyster
- Department of Biophysics and Biochemistry, UCSF, San Francisco, California, USA.,Howard Hughes Medical Institute and.,Department of Microbiology and Immunology, UCSF, San Francisco, California, USA
| | | | - Margot Thome
- Department of Biochemistry, Center of Immunity and Infection, University of Lausanne, Epalinges, Switzerland
| | - Peter C Lucas
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
27
|
Zhang T, Sun J, Cheng J, Yin W, Li J, Miller H, Herrada AA, Gu H, Song H, Chen Y, Gong Q, Liu C. The role of ubiquitinase in B cell development and function. J Leukoc Biol 2020; 109:395-405. [PMID: 32816356 DOI: 10.1002/jlb.1mr0720-185rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 11/10/2022] Open
Abstract
Ubiquitinases are a select group of enzymes that modify target proteins through ubiquitination, which plays a crucial role in the regulation of protein degradation, location, and function. B lymphocytes that originated from bone marrow hematopoietic stem cells (HSC), exert humoral immune functions by differentiating into plasma cells and producing antibodies. Previous studies have shown that ubiquitination is involved in the regulation of the cell cycle and signal transduction important for B lymphocyte development and function. In this review, how ubiquitinases regulate B cell development, activation, apoptosis, and proliferation is discussed, which could help in understanding the physiological processes and diseases related to B cells and also provides potential new targets for further studies.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianxuan Sun
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiali Cheng
- Department of hematology, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yin
- Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingwen Li
- Department of hematology, Wuhan Union Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- Department of Intracellular Pathogens, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Andrés A Herrada
- Lymphatic and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Heng Gu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongmei Song
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 1, Shuaifuyuan, Dongcheng District, Beijing, China
| | - Yan Chen
- The Second Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Wei Z, Zhang Y, Chen J, Hu Y, Jia P, Wang X, Zhao Q, Deng Y, Li N, Zang Y, Qin J, Wang X, Lu W. Pathogenic CARD11 mutations affect B cell development and differentiation through a noncanonical pathway. Sci Immunol 2020; 4:4/41/eaaw5618. [PMID: 31784498 DOI: 10.1126/sciimmunol.aaw5618] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 06/29/2019] [Accepted: 10/24/2019] [Indexed: 01/10/2023]
Abstract
Pathogenic CARD11 mutations cause aberrant nuclear factor κB (NF-κB) activation, which is presumably responsible for multiple immunological disorders. However, whether there is an NF-κB-independent regulatory mechanism contributing to CARD11 mutations related to pathogenesis remains undefined. Using three distinct genetic mouse models, the Card11 knockout (KO) mouse model mimicking primary immunodeficiency, the CARD11 E134G point mutation mouse model representing BENTA (B cell expansion with NF-κB and T cell anergy) disease, and the mouse model bearing oncogenic K215M mutation, we show that CARD11 has a noncanonical function as a negative regulator of the AKT-FOXO1 signal axis, independent of NF-κB activation. Although BENTA disease-related E134G mutant elevates NF-κB activation, we find that E134G mutant mice phenotypically copy Card11 KO mice, in which NF-κB activation is disrupted. Mechanistically, the E134G mutant causes exacerbated AKT activation and reduced FOXO1 protein in B cells similar to that in Card11 KO cells. Moreover, the oncogenic CARD11 mutant K215M reinforces the importance of the noncanonical function of CARD11. In contrast to the E134G mutant, K215M shows a stronger inhibitory effect on AKT activation and more stabilized FOXO1. Likewise, E134G and K215M mutants have converse impacts on B cell development and differentiation. Our results demonstrate that, besides NF-κB, CARD11 also governs the AKT/FOXO1 signaling pathway in B cells. The critical role of CARD11 is further revealed by the effects of pathogenic CARD11 mutants on this noncanonical regulatory function on the AKT-FOXO1 signaling axis.
Collapse
Affiliation(s)
- Zheng Wei
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Zhang
- Division of Immunotherapy, Institute of Human Virology (IHV), School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Jingjing Chen
- Department of Immunology, Nanjing Medical University, 101 Longmain Road, Nanjing 211166, China
| | - Yu Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Pan Jia
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xuelei Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qifang Zhao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yicong Deng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ni Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi Zang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jun Qin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoming Wang
- Department of Immunology, Nanjing Medical University, 101 Longmain Road, Nanjing 211166, China.
| | - Wei Lu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
29
|
Schnappauf O, Aksentijevich I. Mendelian diseases of dysregulated canonical NF-κB signaling: From immunodeficiency to inflammation. J Leukoc Biol 2020; 108:573-589. [PMID: 32678922 DOI: 10.1002/jlb.2mr0520-166r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/05/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022] Open
Abstract
NF-κB is a master transcription factor that activates the expression of target genes in response to various stimulatory signals. Activated NF-κB mediates a plethora of diverse functions including innate and adaptive immune responses, inflammation, cell proliferation, and NF-κB is regulated through interactions with IκB inhibitory proteins, which are in turn regulated by the inhibitor of κB kinase (IKK) complex. Together, these 3 components form the core of the NF-κB signalosomes that have cell-specific functions which are dependent on the interactions with other signaling molecules and pathways. The activity of NF-κB pathway is also regulated by a variety of post-translational modifications including phosphorylation and ubiquitination by Lys63, Met1, and Lys48 ubiquitin chains. The physiologic role of NF-κB is best studied in the immune system due to discovery of many human diseases caused by pathogenic variants in various proteins that constitute the NF-κB pathway. These disease-causing variants can act either as gain-of-function (GoF) or loss-of-function (LoF) and depending on the function of mutated protein, can cause either immunodeficiency or systemic inflammation. Typically, pathogenic missense variants act as GoF and they lead to increased activity in the pathway. LoF variants can be inherited as recessive or dominant alleles and can cause either a decrease or an increase in pathway activity. Dominantly inherited LoF variants often result in haploinsufficiency of inhibitory proteins. Here, we review human Mendelian immunologic diseases, which results from mutations in different molecules in the canonical NF-κB pathway and surprisingly present with a continuum of clinical features including immunodeficiency, atopy, autoimmunity, and autoinflammation.
Collapse
Affiliation(s)
- Oskar Schnappauf
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ivona Aksentijevich
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
30
|
Cheng J, Maurer LM, Kang H, Lucas PC, McAllister-Lucas LM. Critical protein-protein interactions within the CARMA1-BCL10-MALT1 complex: Take-home points for the cell biologist. Cell Immunol 2020; 355:104158. [PMID: 32721634 DOI: 10.1016/j.cellimm.2020.104158] [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: 03/03/2020] [Revised: 05/25/2020] [Accepted: 07/03/2020] [Indexed: 12/24/2022]
Abstract
The CBM complex, which is composed of the proteins CARMA1, BCL10, and MALT1, serves multiple pivotal roles as a mediator of T-cell receptor and B-cell receptor-dependent NF-κB induction and lymphocyte activation. CARMA1, BCL10, and MALT1 are each proto-oncoproteins and dysregulation of CBM signaling, as a result of somatic gain-of-function mutation or chromosomal translocation, is a hallmark of multiple lymphoid malignancies including Activated B-cell Diffuse Large B-cell Lymphoma. Moreover, loss-of-function as well as gain-of-function germline mutations in CBM complex proteins have been associated with a range of immune dysregulation syndromes. A wealth of detailed structural information has become available over the past decade through meticulous interrogation of the interactions between CBM components. Here, we review key findings regarding the biochemical nature of these protein-protein interactions which have ultimately led the field to a sophisticated understanding of how these proteins assemble into high-order filamentous CBM complexes. To date, approaches to therapeutic inhibition of the CBM complex for the treatment of lymphoid malignancy and/or auto-immunity have focused on blocking MALT1 protease function. We also review key studies relating to the structural impact of MALT1 protease inhibitors on key protein-protein interactions.
Collapse
Affiliation(s)
- Jing Cheng
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittburgh, PA, USA
| | - Lisa M Maurer
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittburgh, PA, USA
| | - Heejae Kang
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peter C Lucas
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | |
Collapse
|
31
|
Lu HY, Biggs CM, Blanchard-Rohner G, Fung SY, Sharma M, Turvey SE. Germline CBM-opathies: From immunodeficiency to atopy. J Allergy Clin Immunol 2020; 143:1661-1673. [PMID: 31060714 DOI: 10.1016/j.jaci.2019.03.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/09/2019] [Accepted: 03/15/2019] [Indexed: 12/31/2022]
Abstract
Caspase recruitment domain (CARD) protein-B cell CLL/lymphoma 10 (BCL10)-MALT1 paracaspase (MALT1) [CBM] complexes are critical signaling adaptors that facilitate immune and inflammatory responses downstream of both cell surface and intracellular receptors. Germline mutations that alter the function of members of this complex (termed CBM-opathies) cause a broad array of clinical phenotypes, ranging from profound combined immunodeficiency to B-cell lymphocytosis. With an increasing number of patients being described in recent years, the clinical spectrum of diseases associated with CBM-opathies is rapidly expanding and becoming unexpectedly heterogeneous. Here we review major discoveries that have shaped our understanding of CBM complex biology, and we provide an overview of the clinical presentation, diagnostic approach, and treatment options for those carrying germline mutations affecting CARD9, CARD11, CARD14, BCL10, and MALT1.
Collapse
Affiliation(s)
- Henry Y Lu
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; Experimental Medicine Program, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Catherine M Biggs
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; Experimental Medicine Program, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Geraldine Blanchard-Rohner
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shan-Yu Fung
- Department of Immunology, Tianjin Medical University, Tianjin, China; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin, China
| | - Mehul Sharma
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stuart E Turvey
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; Experimental Medicine Program, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
32
|
Singh M, Jackson KJL, Wang JJ, Schofield P, Field MA, Koppstein D, Peters TJ, Burnett DL, Rizzetto S, Nevoltris D, Masle-Farquhar E, Faulks ML, Russell A, Gokal D, Hanioka A, Horikawa K, Colella AD, Chataway TK, Blackburn J, Mercer TR, Langley DB, Goodall DM, Jefferis R, Gangadharan Komala M, Kelleher AD, Suan D, Rischmueller M, Christ D, Brink R, Luciani F, Gordon TP, Goodnow CC, Reed JH. Lymphoma Driver Mutations in the Pathogenic Evolution of an Iconic Human Autoantibody. Cell 2020; 180:878-894.e19. [PMID: 32059783 DOI: 10.1016/j.cell.2020.01.029] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 11/11/2019] [Accepted: 01/22/2020] [Indexed: 12/26/2022]
Abstract
Pathogenic autoantibodies arise in many autoimmune diseases, but it is not understood how the cells making them evade immune checkpoints. Here, single-cell multi-omics analysis demonstrates a shared mechanism with lymphoid malignancy in the formation of public rheumatoid factor autoantibodies responsible for mixed cryoglobulinemic vasculitis. By combining single-cell DNA and RNA sequencing with serum antibody peptide sequencing and antibody synthesis, rare circulating B lymphocytes making pathogenic autoantibodies were found to comprise clonal trees accumulating mutations. Lymphoma driver mutations in genes regulating B cell proliferation and V(D)J mutation (CARD11, TNFAIP3, CCND3, ID3, BTG2, and KLHL6) were present in rogue B cells producing the pathogenic autoantibody. Antibody V(D)J mutations conferred pathogenicity by causing the antigen-bound autoantibodies to undergo phase transition to insoluble aggregates at lower temperatures. These results reveal a pre-neoplastic stage in human lymphomagenesis and a cascade of somatic mutations leading to an iconic pathogenic autoantibody.
Collapse
Affiliation(s)
- Mandeep Singh
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | | | - Jing J Wang
- Department of Immunology, Flinders University and SA Pathology, Bedford Park, SA 5042, Australia
| | - Peter Schofield
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Matt A Field
- Australian Institute of Tropical Health and Medicine and Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Smithfield, QLD 4878, Australia; The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
| | - David Koppstein
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Timothy J Peters
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Deborah L Burnett
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Simone Rizzetto
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Damien Nevoltris
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Etienne Masle-Farquhar
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Megan L Faulks
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Amanda Russell
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Divya Gokal
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Asami Hanioka
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia; Tokyo Medical and Dental University, Tokyo 113-851, Japan
| | - Keisuke Horikawa
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
| | - Alexander D Colella
- Department of Immunology, Flinders University and SA Pathology, Bedford Park, SA 5042, Australia; Flinders Proteomics Facility, Flinders University, Bedford Park, SA 5042, Australia
| | - Timothy K Chataway
- Flinders Proteomics Facility, Flinders University, Bedford Park, SA 5042, Australia
| | - James Blackburn
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Tim R Mercer
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia; Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
| | - David B Langley
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - D Margaret Goodall
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Roy Jefferis
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | | - Anthony D Kelleher
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Dan Suan
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; Westmead Clinical School, The University of Sydney, Westmead, NSW 2145, Australia
| | - Maureen Rischmueller
- Rheumatology Department, The Queen Elizabeth Hospital and Discipline of Medicine, University of Adelaide, Woodville South, SA 5011, Australia
| | - Daniel Christ
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Robert Brink
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Fabio Luciani
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW 2052, Australia; School of Medical Sciences and Cellular Genomics Futures Institute, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Tom P Gordon
- Department of Immunology, Flinders University and SA Pathology, Bedford Park, SA 5042, Australia
| | - Christopher C Goodnow
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; School of Medical Sciences and Cellular Genomics Futures Institute, UNSW Sydney, Sydney, NSW 2052, Australia.
| | - Joanne H Reed
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia.
| |
Collapse
|
33
|
Shields AM, Bauman BM, Hargreaves CE, Pollard AJ, Snow AL, Patel SY. A Novel, Heterozygous Three Base-Pair Deletion in CARD11 Results in B Cell Expansion with NF-κB and T Cell Anergy Disease. J Clin Immunol 2020; 40:406-411. [PMID: 31897776 DOI: 10.1007/s10875-019-00729-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022]
Abstract
Germline gain-of-function mutations in CARD11 lead to the primary immunodeficiency, B cell expansion with NF-κB, and T cell anergy (BENTA). Herein, we report the case of a girl, presenting at 2 years of age with lymphocytosis and splenomegaly in whom a novel, in-frame, three base pair deletion in CARD11 was identified resulting in the deletion of a single lysine residue (K215del) from the coiled-coil domain. In vitro functional assays demonstrated that this variant leads to a subtle increase in baseline NF-κB signaling and impaired proliferative responses following T cell receptor and mitogenic stimulation. Previously reported immunological defects associated with BENTA appear mild in our patient who is now 6 years of age; a B cell lymphocytosis and susceptibility to upper respiratory tract infections persist; however, she has broad, sustained responses to protein-polysaccharide conjugate vaccines and displays normal proliferative responses to ex vivo T cell stimulation.
Collapse
Affiliation(s)
- Adrian M Shields
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, UK.
| | - Bradly M Bauman
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Chantal E Hargreaves
- Department of Clinical Immunology, John Radcliffe Hospital, Oxford, OX3 9DU, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Smita Y Patel
- Department of Clinical Immunology, John Radcliffe Hospital, Oxford, OX3 9DU, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
34
|
Xu L, He J, Cai Q, Li M, Pu X, Guo Y. An effective seven-CpG-based signature to predict survival in renal clear cell carcinoma by integrating DNA methylation and gene expression. Life Sci 2020; 243:117289. [PMID: 31926254 DOI: 10.1016/j.lfs.2020.117289] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/29/2019] [Accepted: 01/07/2020] [Indexed: 12/16/2022]
Abstract
AIMS Currently, using clinicopathological risk factors only is not far from effective to evaluate the risk of disease progression in renal clear cell carcinoma (KIRC) patients. Molecular biomarkers might improve risk stratification of KIRC. DNA methylation occurs the whole process of tumor development and transcriptional disorders are also one of the important characteristics of tumor. Hence, this study aims to develop an effective and independent prognostic signature for KIRC patients by Integrating DNA methylation and gene expression. MAIN METHODS Difference analysis was conducted on DNA methylation sites and gene expression data. The Spearman's rank correlation and univariate Cox regression analysis were used to screen out the CpG sites that related with RNAs' expression and KIRC patients' overall survival. Then, a five-CpG-based prognostic classifier was established using LASSO Cox regression method. KEY FINDINGS The seven-CpG-based classifier can successfully divide KIRC patients into high-risk from low-risk groups, even after adjustment for standard clinical prognostic factors, such as age, stage, gender and grade. Moreover, the seven-CpG-based signature was more effective as independent prognostic factors than the combined model of these clinical factors. Six differential mRNA genes corresponding to the seven CpG sites are all related to human cancers by functional exploration. The gene functional and pathway enrichment analysis found that genes in immune-related pathways were remarkably different in high and low-risk groups. SIGNIFICANCE The new seven-CpG-based signature could helpfully provide insights into the underlying mechanism of KIRC and may be a powerful independent biomarker for predicting of the survival of KIRC patients.
Collapse
Affiliation(s)
- Lei Xu
- College of Chemistry, Sichuan University, Chengdu, Sichuan, China
| | - Jian He
- College of Chemistry, Sichuan University, Chengdu, Sichuan, China
| | - Qihang Cai
- College of Chemistry, Sichuan University, Chengdu, Sichuan, China
| | - Menglong Li
- College of Chemistry, Sichuan University, Chengdu, Sichuan, China
| | - Xuemei Pu
- College of Chemistry, Sichuan University, Chengdu, Sichuan, China
| | - Yanzhi Guo
- College of Chemistry, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
35
|
Ma J, Fu L, Gu H, Chen Z, Zhang J, Zhao S, Zhu X, Liu H, Wu R. Screening for Genetic Mutations for the Early Diagnosis of Common Variable Immunodeficiency in Children With Refractory Immune Thrombocytopenia: A Retrospective Data Analysis From a Tertiary Children's Center. Front Pediatr 2020; 8:595135. [PMID: 33425813 PMCID: PMC7793988 DOI: 10.3389/fped.2020.595135] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 11/12/2020] [Indexed: 12/19/2022] Open
Abstract
Aim: This study aimed to identify common variable immunodeficiency (CVID) by high-throughput next-generation sequencing (NGS) in children with refractory immune thrombocytopenia (RITP) to facilitate early diagnosis. Methods: CVID-related genetic mutations were explored in patients with RITP during 2016-2019. They were tested consecutively through NGS by the ITP team of the tertiary children hospital in China. An evaluation system was devised based on the phenotype, genetic rule, and serum immunoglobulins (Igs) of all patients with RITP. The patients were divided into highly suspicious, suspicious, and negative groups using the evaluation system. Results: Among 176 patients with RITP, 16 (9.1%) harbored CVID-related genetic mutations: 8 (4.5%) were highly suspicious of CVIDs. Five had mutations in tumor necrosis factor receptor superfamily 13B (TNFRSF13B), one in lipopolysaccharide responsive beige-like anchor protein (LRBA), one in nuclear factor kappa-B2 (NF-κB2), and one in caspase recruitment domain11 (CARD11). Others were classified into the suspicious group because the clinical phenotype and pedigree were suggestive, yet insufficient, for diagnosis. Repeated infection existed in all patients. Two had an allergic disease. Positive autoimmune serologies were noted in 62.5%. Five had a definite positive family history. The median serum immunoglobulin (Ig)A, IgG, and IgM levels were 0.3875, 6.14, and 0.522 g/L, respectively. Nearly 85.7% of patients had insufficient serum IgA levels, while 37.5% had low IgG and IgM levels. Conclusions: High-throughput NGS and a thorough review of the medical history are beneficial for the early diagnosis of patients without any significant clinical characteristics, distinguishing them from those with primary pediatric ITP. The cases suspicious of CVID need further investigation and follow-up to avoid deterioration.
Collapse
Affiliation(s)
- Jingyao Ma
- Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Oncology Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China.,National Key Discipline of Pediatrics, Capital Medical University, Beijing, China.,Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Lingling Fu
- Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Oncology Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China.,National Key Discipline of Pediatrics, Capital Medical University, Beijing, China.,Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Hao Gu
- Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Oncology Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China.,National Key Discipline of Pediatrics, Capital Medical University, Beijing, China.,Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Zhenping Chen
- Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Oncology Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China.,National Key Discipline of Pediatrics, Capital Medical University, Beijing, China.,Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Jialu Zhang
- Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Oncology Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China.,National Key Discipline of Pediatrics, Capital Medical University, Beijing, China.,Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Shasha Zhao
- Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Oncology Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China.,National Key Discipline of Pediatrics, Capital Medical University, Beijing, China.,Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Xiaojing Zhu
- Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Oncology Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China.,National Key Discipline of Pediatrics, Capital Medical University, Beijing, China.,Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Huiqing Liu
- Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Oncology Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China.,National Key Discipline of Pediatrics, Capital Medical University, Beijing, China.,Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Runhui Wu
- Beijing Key Laboratory of Pediatric Hematology Oncology, Hematology Oncology Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China.,National Key Discipline of Pediatrics, Capital Medical University, Beijing, China.,Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| |
Collapse
|
36
|
Wang Z, Hutcherson SM, Yang C, Jattani RP, Tritapoe JM, Yang YK, Pomerantz JL. Coordinated regulation of scaffold opening and enzymatic activity during CARD11 signaling. J Biol Chem 2019; 294:14648-14660. [PMID: 31391255 PMCID: PMC6779434 DOI: 10.1074/jbc.ra119.009551] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/01/2019] [Indexed: 11/06/2022] Open
Abstract
The activation of key signaling pathways downstream of antigen receptor engagement is critically required for normal lymphocyte activation during the adaptive immune response. CARD11 is a multidomain signaling scaffold protein required for antigen receptor signaling to NF-κB, c-Jun N-terminal kinase, and mTOR. Germline mutations in the CARD11 gene result in at least four types of primary immunodeficiency, and somatic CARD11 gain-of-function mutations drive constitutive NF-κB activity in diffuse large B cell lymphoma and other lymphoid cancers. In response to antigen receptor triggering, CARD11 transitions from a closed, inactive state to an open, active scaffold that recruits multiple signaling partners into a complex to relay downstream signaling. However, how this signal-induced CARD11 conversion occurs remains poorly understood. Here we investigate the role of Inducible Element 1 (IE1), a short regulatory element in the CARD11 Inhibitory Domain, in the CARD11 signaling cycle. We find that IE1 controls the signal-dependent Opening Step that makes CARD11 accessible to the binding of cofactors, including Bcl10, MALT1, and the HOIP catalytic subunit of the linear ubiquitin chain assembly complex. Surprisingly, we find that IE1 is also required at an independent step for the maximal activation of HOIP and MALT1 enzymatic activity after cofactor recruitment to CARD11. This role of IE1 reveals that there is an Enzymatic Activation Step in the CARD11 signaling cycle that is distinct from the Cofactor Association Step. Our results indicate that CARD11 has evolved to actively coordinate scaffold opening and the induction of enzymatic activity among recruited cofactors during antigen receptor signaling.
Collapse
Affiliation(s)
- Zhaoquan Wang
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Shelby M Hutcherson
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Chao Yang
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Rakhi P Jattani
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Julia M Tritapoe
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Yong-Kang Yang
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Joel L Pomerantz
- Department of Biological Chemistry and Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
37
|
Sloboda N, Sorlin A, Valduga M, Beri-Dexheimer M, Bilbault C, Fouyssac F, Becker A, Lambert L, Bonnet C, Leheup B. Deletion of chr7p22 and chr15q11: Two Familial Cases of Immune Deficiency: Extending the Phenotype Toward Dysimmunity. Front Immunol 2019; 10:1871. [PMID: 31474980 PMCID: PMC6707040 DOI: 10.3389/fimmu.2019.01871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/24/2019] [Indexed: 11/29/2022] Open
Abstract
Background: We report here two new familial cases of associated del15q11 and del7p22, with the latter underlining the clinical variability of this deletion. Two siblings patients presented a similar familial imbalanced translocation, originating from a balanced maternal translocation, with deletions of 7p22 and of 15q11 [arr[GRCh37] 7p22.3-p22.2(42976-3736851)x1, 15q11.1-q11.2(20172544-24979427)x1]. Methods: We used aCGH array, FISH, and karyotype for studying the phenotype of the two patients. Results: The 7p22 deletion (3.5 Mb) contained 58 genes, including several OMIM genes. Patients 1 and 2 exhibited acquisition delays, morphological particularities, and hypogammaglobulinemia, which was more severe in patient 1. Patient 1 presented also with cerebral vasculitis. Conclusion: We discuss here how the PDGFa, CARD11, LFNG, GPER1, and MAFK genes, included in the deletion 7p22, could be involved in the clinical and biological features of the two patients.
Collapse
Affiliation(s)
- Natacha Sloboda
- Clinic Genetics Department, Children Hospital, CHRU Nancy, Nancy, France
| | - Arthur Sorlin
- Clinic Genetics Department, Children Hospital, CHRU Nancy, Nancy, France
| | | | | | - Claire Bilbault
- Infantile Medicine Department (Neuropediatrics), Children Hospital, CHRU Nancy, Nancy, France
| | - Fanny Fouyssac
- Infantile Medicine Department (Hematopediatrics), Children Hospital, CHRU Nancy, Nancy, France
| | | | - Laëtitia Lambert
- Clinic Genetics Department, Children Hospital, CHRU Nancy, Nancy, France
| | | | - Bruno Leheup
- Clinic Genetics Department, Children Hospital, CHRU Nancy, Nancy, France
| |
Collapse
|
38
|
Sacco KA, Milner JD. Gene-environment interactions in primary atopic disorders. Curr Opin Immunol 2019; 60:148-155. [PMID: 31302571 DOI: 10.1016/j.coi.2019.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/14/2019] [Indexed: 12/18/2022]
Abstract
Environmental factors modify disease presentation and severity in allergic disorders. Primary atopic disorders (PADs) are a heterogenous group of single gene disorders that lead to significant atopic and allergic disease manifestations. However, a number of these monogenic diseases have variable penetrance suggesting that gene-gene and/or gene-environment interactions could modulate the clinical phenotype. Environmental factors such as diet, the microbiome at the epithelial-environment interface, the presence and/or extent of infection, and psychologic stress can alter disease phenotypic expression of allergic diseases, and PADs provide discrete contexts in which to understand these influences. We outline how gene-environment interactions likely contribute to a variable penetrance and expressivity in PADs. Dietary modifications of both macronutrients and/or micronutrients alter T-cell metabolism and may influence effector T-cell function. The mucosal microbiome may affect local inflammation and may remotely influence regulatory elements, while psychologic stress can affect mast cell and other allergic effector cell function. Understanding gene-environment interactions in PADs can hopefully provide a foundation for interrogating gene-environment interactions to common allergic disorders, and also present opportunities for personalized interventions based on the altered pathways and environmental influences in affected individuals.
Collapse
Affiliation(s)
- Keith A Sacco
- Laboratory of Allergic Diseases, NIAID, NIH, 9000 Rockville Pike, NIH Building 10 Room 11N240A, United States
| | - Joshua D Milner
- Laboratory of Allergic Diseases, NIAID, NIH, 9000 Rockville Pike, NIH Building 10 Room 11N240A, United States.
| |
Collapse
|
39
|
Policheni A, Horikawa K, Milla L, Kofler J, Bouillet P, Belz GT, O'Reilly LA, Goodnow CC, Strasser A, Gray DHD. CARD11 is dispensable for homeostatic responses and suppressive activity of peripherally induced FOXP3
+
regulatory T cells. Immunol Cell Biol 2019; 97:740-752. [DOI: 10.1111/imcb.12268] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 05/12/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Antonia Policheni
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Keisuke Horikawa
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics The John Curtin School of Medical Research The Australian National University Canberra ACT Australia
| | - Liz Milla
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Jennifer Kofler
- Australian Cancer Research Foundation Department of Cancer Biology and Therapeutics The John Curtin School of Medical Research The Australian National University Canberra ACT Australia
| | - Philippe Bouillet
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Gabrielle T Belz
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Lorraine A O'Reilly
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | | | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Daniel HD Gray
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| |
Collapse
|
40
|
Lork M, Staal J, Beyaert R. Ubiquitination and phosphorylation of the CARD11-BCL10-MALT1 signalosome in T cells. Cell Immunol 2018; 340:103877. [PMID: 30514565 DOI: 10.1016/j.cellimm.2018.11.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/02/2018] [Indexed: 12/16/2022]
Abstract
Antigen receptor-induced signaling plays an important role in inflammation and immunity. Formation of a CARD11-BCL10-MALT1 (CBM) signaling complex is a key event in T- and B cell receptor-induced gene expression by regulating NF-κB activation and mRNA stability. Deregulated CARD11, BCL10 or MALT1 expression or CBM signaling have been associated with immunodeficiency, autoimmunity and cancer, indicating that CBM formation and function have to be tightly regulated. Over the past years great progress has been made in deciphering the molecular mechanisms of assembly and disassembly of the CBM complex. In this context, several posttranslational modifications play an indispensable role in regulating CBM function and downstream signal transduction. In this review we summarize how the different CBM components as well as their interplay are regulated by protein ubiquitination and phosphorylation in the context of T cell receptor signaling.
Collapse
Affiliation(s)
- Marie Lork
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Unit of Molecular Signal Transduction in Inflammation, Center for Inflammation Research, VIB, Ghent, Belgium
| | - Jens Staal
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Unit of Molecular Signal Transduction in Inflammation, Center for Inflammation Research, VIB, Ghent, Belgium
| | - Rudi Beyaert
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Unit of Molecular Signal Transduction in Inflammation, Center for Inflammation Research, VIB, Ghent, Belgium.
| |
Collapse
|
41
|
Ruland J, Hartjes L. CARD–BCL-10–MALT1 signalling in protective and pathological immunity. Nat Rev Immunol 2018; 19:118-134. [DOI: 10.1038/s41577-018-0087-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
42
|
Seeholzer T, Kurz S, Schlauderer F, Woods S, Gehring T, Widmann S, Lammens K, Krappmann D. BCL10-CARD11 Fusion Mimics an Active CARD11 Seed That Triggers Constitutive BCL10 Oligomerization and Lymphocyte Activation. Front Immunol 2018; 9:2695. [PMID: 30515170 PMCID: PMC6255920 DOI: 10.3389/fimmu.2018.02695] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/31/2018] [Indexed: 12/16/2022] Open
Abstract
Assembly of the CARD11/CARMA1-BCL10-MALT1 (CBM) signaling complex upon T or B cell antigen receptor (TCR or BCR) engagement drives lymphocyte activation. Recruitment of pre-assembled BCL10-MALT1 complexes to CARD11 fosters activation of the MALT1 protease and canonical NF-κB signaling. Structural data and in vitro assays have suggested that CARD11 acts as a seed that nucleates the assembly of BCL10 filaments, but the relevance of these findings for CBM complex assembly in cells remains unresolved. To uncouple cellular CARD11 recruitment of BCL10 and BCL10 filament assembly, we generated a BCL10-CARD11 fusion protein that links the C-terminus of BCL10 to the N-terminus of CARD11. When stably expressed in CARD11 KO Jurkat T cells, the BCL10-CARD11 fusion induced constitutive MALT1 activation. Furthermore, in CARD11 KO BJAB B cells, BCL10-CARD11 promoted constitutive NF-κB activation to a similar extent as CARD11 containing oncogenic driver mutations. Using structure-guided destructive mutations in the CARD11-BCL10 (CARD11 R35A) or BCL10-BCL10 (BCL10 R42E) interfaces, we demonstrate that chronic activation by the BCL10-CARD11 fusion protein was independent of the CARD11 CARD. However, activation strictly relied upon the ability of the BCL10 CARD to form oligomers. Thus, by combining distinct CARD mutations in the context of constitutively active BCL10-CARD11 fusion proteins, we provide evidence that BCL10-MALT1 recruitment to CARD11 and BCL10 oligomerization are interconnected processes, which bridge the CARD11 seed to downstream pathways in lymphocytes.
Collapse
Affiliation(s)
- Thomas Seeholzer
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Susanne Kurz
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Simone Woods
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Torben Gehring
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Simon Widmann
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Katja Lammens
- Gene Center, Ludwig-Maximilians University, Munich, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
43
|
Zotti T, Polvere I, Voccola S, Vito P, Stilo R. CARD14/CARMA2 Signaling and its Role in Inflammatory Skin Disorders. Front Immunol 2018; 9:2167. [PMID: 30319628 PMCID: PMC6168666 DOI: 10.3389/fimmu.2018.02167] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/03/2018] [Indexed: 12/23/2022] Open
Abstract
CARMA proteins represent a family of scaffold molecules which play several crucial biological functions, including regulation of immune response and inflammation, tissue homeostasis, and modulation of G-Protein Coupled Receptor (GPCR) signaling. Among the CARMA proteins, CARD14/CARMA2 and its alternatively spliced isoforms are specifically expressed in epithelial cells and keratinocytes. Recent evidences have shown that CARD14/CARMA2 mediates induction of inflammatory response in keratinocytes, and that mutations in CARD14/CARMA2 gene segregate with familial transmission of chronic inflammatory disorders of the human skin. Similarly to CARD11/CARMA1 and CARD10/CARMA3, CARD14/CARMA2 signaling occurs trough formation of a trimeric complex which includes BCL10 and MALT1 proteins. However, it is becoming increasingly evident that in addition to the CBM complex components, a number of accessory molecules are able to finely modulate the signals conveyed on and amplified by CARD14/CARMA2. The study of these molecules is important both to understand the molecular mechanisms that underlie the role of CARMA2 in keratinocytes and because they represent potential therapeutic targets for the development of therapeutic strategies aiming at the treatment of inflammatory diseases of the human skin. In this review, we provide an overview on the molecular mechanisms mediating CARD14/CARMA2 signaling and its implication in our understanding of the pathogenesis of human inflammatory skin disorders.
Collapse
Affiliation(s)
- Tiziana Zotti
- Genus Biotechnology, Università degli Studi del Sannio, Benevento, Italy.,Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, Benevento, Italy
| | - Immacolata Polvere
- Genus Biotechnology, Università degli Studi del Sannio, Benevento, Italy.,Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, Benevento, Italy
| | - Serena Voccola
- Genus Biotechnology, Università degli Studi del Sannio, Benevento, Italy.,Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, Benevento, Italy
| | - Pasquale Vito
- Genus Biotechnology, Università degli Studi del Sannio, Benevento, Italy.,Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, Benevento, Italy
| | - Romania Stilo
- Genus Biotechnology, Università degli Studi del Sannio, Benevento, Italy.,Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, Benevento, Italy
| |
Collapse
|
44
|
Bedsaul JR, Carter NM, Deibel KE, Hutcherson SM, Jones TA, Wang Z, Yang C, Yang YK, Pomerantz JL. Mechanisms of Regulated and Dysregulated CARD11 Signaling in Adaptive Immunity and Disease. Front Immunol 2018; 9:2105. [PMID: 30283447 PMCID: PMC6156143 DOI: 10.3389/fimmu.2018.02105] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 08/28/2018] [Indexed: 01/02/2023] Open
Abstract
CARD11 functions as a key signaling scaffold that controls antigen-induced lymphocyte activation during the adaptive immune response. Somatic mutations in CARD11 are frequently found in Non-Hodgkin lymphoma, and at least three classes of germline CARD11 mutations have been described as the basis for primary immunodeficiency. In this review, we summarize our current understanding of how CARD11 signals, how its activity is regulated, and how mutations bypass normal regulation to cause disease.
Collapse
Affiliation(s)
- Jacquelyn R Bedsaul
- Department of Biological Chemistry, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nicole M Carter
- Department of Biological Chemistry, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Katelynn E Deibel
- Department of Biological Chemistry, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shelby M Hutcherson
- Department of Biological Chemistry, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Tyler A Jones
- Department of Biological Chemistry, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Zhaoquan Wang
- Department of Biological Chemistry, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Chao Yang
- Department of Biological Chemistry, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yong-Kang Yang
- Department of Biological Chemistry, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Joel L Pomerantz
- Department of Biological Chemistry, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
45
|
Lu HY, Bauman BM, Arjunaraja S, Dorjbal B, Milner JD, Snow AL, Turvey SE. The CBM-opathies-A Rapidly Expanding Spectrum of Human Inborn Errors of Immunity Caused by Mutations in the CARD11-BCL10-MALT1 Complex. Front Immunol 2018; 9:2078. [PMID: 30283440 PMCID: PMC6156466 DOI: 10.3389/fimmu.2018.02078] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 08/22/2018] [Indexed: 01/06/2023] Open
Abstract
The caspase recruitment domain family member 11 (CARD11 or CARMA1)-B cell CLL/lymphoma 10 (BCL10)-MALT1 paracaspase (MALT1) [CBM] signalosome complex serves as a molecular bridge between cell surface antigen receptor signaling and the activation of the NF-κB, JNK, and mTORC1 signaling axes. This positions the CBM complex as a critical regulator of lymphocyte activation, proliferation, survival, and metabolism. Inborn errors in each of the CBM components have now been linked to a diverse group of human primary immunodeficiency diseases termed "CBM-opathies." Clinical manifestations range from severe combined immunodeficiency to selective B cell lymphocytosis, atopic disease, and specific humoral defects. This surprisingly broad spectrum of phenotypes underscores the importance of "tuning" CBM signaling to preserve immune homeostasis. Here, we review the distinct clinical and immunological phenotypes associated with human CBM complex mutations and introduce new avenues for targeted therapeutic intervention.
Collapse
Affiliation(s)
- Henry Y Lu
- Department of Pediatrics, British Columbia Children's Hospital, The University of British Columbia, Vancouver, BC, Canada.,Experimental Medicine Program, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Bradly M Bauman
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Swadhinya Arjunaraja
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Batsukh Dorjbal
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Joshua D Milner
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Stuart E Turvey
- Department of Pediatrics, British Columbia Children's Hospital, The University of British Columbia, Vancouver, BC, Canada.,Experimental Medicine Program, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
46
|
Lu HY, Sharma M, Biggs CM, Huang YH, Shopsowitz KE, Frosk P, Priatel JJ, Rubin TS, Turvey SE. The importance of functional validation after next-generation sequencing: evaluation of a novel CARD11 variant. Pediatr Allergy Immunol 2018; 29:663-668. [PMID: 29808493 DOI: 10.1111/pai.12930] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Henry Y Lu
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada.,Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Mehul Sharma
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Catherine M Biggs
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada.,Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yu-Hsuan Huang
- Department of Pathology and Laboratory Medicine, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Kevin E Shopsowitz
- Department of Medical Genetics, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Patrick Frosk
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada.,Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - John J Priatel
- Department of Pathology and Laboratory Medicine, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Tamar S Rubin
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada.,Division of Allergy and Clinical Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Stuart E Turvey
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada.,Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
47
|
Gehring T, Seeholzer T, Krappmann D. BCL10 - Bridging CARDs to Immune Activation. Front Immunol 2018; 9:1539. [PMID: 30022982 PMCID: PMC6039553 DOI: 10.3389/fimmu.2018.01539] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/21/2018] [Indexed: 11/25/2022] Open
Abstract
Since the B-cell lymphoma/leukemia 10 (BCL10) protein was first described in 1999, numerous studies have elucidated its key functions in channeling adaptive and innate immune signaling downstream of CARMA/caspase-recruitment domain (CARD) scaffold proteins. While T and B cell antigen receptor (TCR/BCR) signaling induces the recruitment of BCL10 bound to mucosa-associated lymphoid tissue (MALT)1 to the lymphocyte-specific CARMA1/CARD11–BCL10–MALT1 (CBM-1) signalosome, alternative CBM complexes utilize different CARMA/CARD scaffolds in distinct innate or inflammatory pathways. BCL10 constitutes the smallest subunit in all CBM signalosomes, containing a 233 amino acid coding for N-terminal CARD as well as a C-terminal Ser/Thr-rich region. BCL10 forms filaments, thereby aggregating into higher-order clusters that mediate and amplify stimulation-induced signals, ultimately leading to MALT1 protease activation and canonical NF-κB and JNK signaling. BCL10 additionally undergoes extensive post-translational regulation involving phosphorylation, ubiquitination, MALT1-catalyzed cleavage, and degradation. Through these feedback and feed-forward events, BCL10 integrates positive and negative regulatory processes that govern the function as well as the dynamic assembly, disassembly, and destruction of CBM complexes. Thus, BCL10 is a critical regulator for activation as well as termination of immune cell signaling, revealing that its role extends far beyond that of a mere linking factor in CBM complexes.
Collapse
Affiliation(s)
- Torben Gehring
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Seeholzer
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
48
|
Kuwabara T, Matsui Y, Ishikawa F, Kondo M. Regulation of T-Cell Signaling by Post-Translational Modifications in Autoimmune Disease. Int J Mol Sci 2018. [PMID: 29534522 PMCID: PMC5877680 DOI: 10.3390/ijms19030819] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The adaptive immune system involves antigen-specific host defense mechanisms mediated by T and B cells. In particular, CD4+ T cells play a central role in the elimination of pathogens. Immunological tolerance in the thymus regulates T lymphocytes to avoid self-components, including induction of cell death in immature T cells expressing the self-reactive T-cell receptor repertoire. In the periphery, mature T cells are also regulated by tolerance, e.g., via induction of anergy or regulatory T cells. Thus, T cells strictly control intrinsic signal transduction to prevent excessive responses or self-reactions. If the inhibitory effects of T cells on these mechanisms are disrupted, T cells may incorrectly attack self-components, which can lead to autoimmune disease. The functions of T cells are supported by post-translational modifications, particularly phosphorylation, of signaling molecules, the proper regulation of which is controlled by endogenous mechanisms within the T cells themselves. In recent years, molecular targeted agents against kinases have been developed for treatment of autoimmune diseases. In this review, we discuss T-cell signal transduction in autoimmune disease and provide an overview of acetylation-mediated regulation of T-cell signaling pathways.
Collapse
Affiliation(s)
- Taku Kuwabara
- Department of Molecular Immunology, Toho University School of Medicine, 5-21-16 Omori-Nishi, Ota-ku, Tokyo 143-8540, Japan.
| | - Yukihide Matsui
- Department of Molecular Immunology, Toho University School of Medicine, 5-21-16 Omori-Nishi, Ota-ku, Tokyo 143-8540, Japan.
| | - Fumio Ishikawa
- Department of Molecular Immunology, Toho University School of Medicine, 5-21-16 Omori-Nishi, Ota-ku, Tokyo 143-8540, Japan.
| | - Motonari Kondo
- Department of Molecular Immunology, Toho University School of Medicine, 5-21-16 Omori-Nishi, Ota-ku, Tokyo 143-8540, Japan.
| |
Collapse
|
49
|
Arjunaraja S, Angelus P, Su HC, Snow AL. Impaired Control of Epstein-Barr Virus Infection in B-Cell Expansion with NF-κB and T-Cell Anergy Disease. Front Immunol 2018; 9:198. [PMID: 29472930 PMCID: PMC5809398 DOI: 10.3389/fimmu.2018.00198] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 01/23/2018] [Indexed: 11/13/2022] Open
Abstract
B-cell expansion with NF-κB and T-cell anergy (BENTA) disease is a B-cell-specific lymphoproliferative disorder caused by germline gain-of-function mutations in CARD11. These mutations force the CARD11 scaffold into an open conformation capable of stimulating constitutive NF-κB activation in lymphocytes, without requiring antigen receptor engagement. Many BENTA patients also suffer from recurrent infections, with 7 out of 16 patients exhibiting chronic, low-grade Epstein–Barr virus (EBV) viremia. In this mini-review, we discuss EBV infection in the pathogenesis and clinical management of BENTA disease, and speculate on mechanisms that could explain inadequate control of viral infection in BENTA patients.
Collapse
Affiliation(s)
- Swadhinya Arjunaraja
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Pamela Angelus
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Clinical Research Directorate/Clinical Monitoring Research Program, Leidos Biomedical Research, Inc., National Cancer Institute at Frederick, Frederick, MD, United States
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
50
|
Elian FA, Yan E, Walter MA. FOXC1, the new player in the cancer sandbox. Oncotarget 2018; 9:8165-8178. [PMID: 29487724 PMCID: PMC5814291 DOI: 10.18632/oncotarget.22742] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 10/28/2017] [Indexed: 01/01/2023] Open
Abstract
In recent years, rapidly accumulating evidence implicates forkhead box C1 (FOXC1) in cancer, especially in studies of basal-like breast cancer (BLBC). Other studies have followed suit, demonstrating that FOXC1 is not only a major player in this breast cancer subtype, but also in hepatocellular carcinoma (HCC), endometrial cancer, Hodgkin's lymphoma (HL), and non-Hodgkin's lymphoma (NHL). The FOXC1 gene encodes a transcription factor that is crucial to mesodermal, neural crest, and ocular development, and mutations found in FOXC1 have been found to cause dominantly inherited Axenfeld-Rieger Syndrome (ARS). Interestingly, while FOXC1 missense mutations that are associated with ARS usually reduce gene activity, increased FOXC1 function now appears to be often linked to more aggressive cancer phenotypes in BLBC, HCC, HL, and NHL. This review discusses not only the role of FOXC1 in cancer cell progression, proliferation, differentiation, and metastasis, but also the underlying mechanisms of how FOXC1 can contribute to aggressive cancer phenotypes.
Collapse
Affiliation(s)
- Fahed A. Elian
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Elizabeth Yan
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Michael A. Walter
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|