1
|
Olivier JF, Langlais D, Jeyakumar T, Polyak MJ, Galarneau L, Cayrol R, Jiang H, Molloy KR, Xu G, Suzuki H, LaCava J, Gros P, Fodil N. CCDC88B interacts with RASAL3 and ARHGEF2 and regulates dendritic cell function in neuroinflammation and colitis. Commun Biol 2024; 7:77. [PMID: 38200184 PMCID: PMC10781698 DOI: 10.1038/s42003-023-05751-9] [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: 09/22/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
CCDC88B is a risk factor for several chronic inflammatory diseases in humans and its inactivation causes a migratory defect in DCs in mice. CCDC88B belongs to a family of cytoskeleton-associated scaffold proteins that feature protein:protein interaction domains. Here, we identified the Rho/Rac Guanine Nucleotide Exchange Factor 2 (ARHGEF2) and the RAS Protein Activator Like 3 (RASAL3) as CCDC88B physical and functional interactors. Mice defective in Arhgef2 or Rasal3 show dampened neuroinflammation, and display altered cellular response and susceptibility to colitis; ARHGEF2 maps to a human Chromosome 1 locus associated with susceptibility to IBD. Arhgef2 and Rasal3 mutant DCs show altered migration and motility in vitro, causing either reduced (Arhgef2) or enhanced (Rasal3) migratory properties. The CCDC88B/RASAL3/ARHGEF2 complex appears to regulate DCs migration by modulating activation of RHOA, with ARHGEF2 and RASAL3 acting in opposite regulatory fashions, providing a molecular mechanism for the involvement of these proteins in DCs immune functions.
Collapse
Affiliation(s)
- Jean-Frederic Olivier
- Department of Biochemistry, McGill University, Montreal, QC, Canada
- McGill Research Center on Complex Traits, Montreal, QC, Canada
| | - David Langlais
- McGill Research Center on Complex Traits, Montreal, QC, Canada
- Department of Human Genetics, Victor Phillip Dahdaleh Institute of Genomic Medicine, Montreal, QC, Canada
| | - Thiviya Jeyakumar
- Department of Biochemistry, McGill University, Montreal, QC, Canada
- McGill Research Center on Complex Traits, Montreal, QC, Canada
| | - Maria J Polyak
- Department of Biochemistry, McGill University, Montreal, QC, Canada
- McGill Research Center on Complex Traits, Montreal, QC, Canada
| | - Luc Galarneau
- Department of Medicine, Sherbrooke University, Sherbrooke, QC, Canada
| | - Romain Cayrol
- Department of Pathology, University of Montreal Hospital Center (CHUM), Montreal, QC, Canada
- University of Montreal Hospital Center Research Center (CR-CHUM), Montreal, QC, Canada
- Department of Pathology and Cellular Biology, University of Montreal, Montreal, QC, Canada
| | - Hua Jiang
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY, USA
| | - Kelly R Molloy
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY, USA
| | - Guoyue Xu
- Department of Human Genetics, Victor Phillip Dahdaleh Institute of Genomic Medicine, Montreal, QC, Canada
| | - Harumi Suzuki
- Department of Immunology and Pathology, National Center for Global Health and Medicine, Tokyo, Japan
| | - John LaCava
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY, USA
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, Groningen, The Netherlands
| | - Philippe Gros
- Department of Biochemistry, McGill University, Montreal, QC, Canada.
- McGill Research Center on Complex Traits, Montreal, QC, Canada.
| | - Nassima Fodil
- McGill Research Center on Complex Traits, Montreal, QC, Canada
- CERMO-FC, Pavillon des Sciences Biologiques, Montreal, QC, Canada
| |
Collapse
|
2
|
Gautam N, Wojciech L, Yap J, Chua YL, Ding EM, Sim DC, Tan AS, Ahl PJ, Prasad M, Tung DW, Connolly JE, Adriani G, Brzostek J, Gascoigne NR. Themis controls T cell activation, effector functions, and metabolism of peripheral CD8 + T cells. Life Sci Alliance 2023; 6:e202302156. [PMID: 37739454 PMCID: PMC10517225 DOI: 10.26508/lsa.202302156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 09/24/2023] Open
Abstract
Themis is important in regulating positive selection of thymocytes during T cell development, but its role in peripheral T cells is less understood. Here, we investigated T cell activation and its sequelae using a tamoxifen-mediated, acute Themis deletion mouse model. We find that proliferation, effector functions including anti-tumor killing, and up-regulation of energy metabolism are severely compromised. This study reveals the phenomenon of peripheral adaptation to loss of Themis, by demonstrating direct TCR-induced defects after acute deletion of Themis that were not evident in peripheral T cells chronically deprived of Themis in dLck-Cre deletion model. Peripheral adaptation to long-term loss was compared using chronic versus acute tamoxifen-mediated deletion and with the (chronic) dLck-Cre deletion model. We found that upon chronic tamoxifen-mediated Themis deletion, there was modulation in the gene expression profile for both TCR and cytokine signaling pathways. This profile overlapped with (chronic) dLck-Cre deletion model. Hence, we found that peripheral adaptation induced changes to both TCR and cytokine signaling modules. Our data highlight the importance of Themis in the activation of CD8+ T cells.
Collapse
Affiliation(s)
- Namrata Gautam
- Translational Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lukasz Wojciech
- Translational Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jiawei Yap
- Translational Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yen Leong Chua
- Translational Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Eyan Mw Ding
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Don Cn Sim
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Alrina Sm Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Patricia J Ahl
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Mukul Prasad
- Translational Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Desmond Wh Tung
- Translational Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - John E Connolly
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Giulia Adriani
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Joanna Brzostek
- Translational Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nicholas Rj Gascoigne
- Translational Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Translational Cancer Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
3
|
Hadjilaou A, Brandi J, Riehn M, Friese MA, Jacobs T. Pathogenetic mechanisms and treatment targets in cerebral malaria. Nat Rev Neurol 2023; 19:688-709. [PMID: 37857843 DOI: 10.1038/s41582-023-00881-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2023] [Indexed: 10/21/2023]
Abstract
Malaria, the most prevalent mosquito-borne infectious disease worldwide, has accompanied humanity for millennia and remains an important public health issue despite advances in its prevention and treatment. Most infections are asymptomatic, but a small percentage of individuals with a heavy parasite burden develop severe malaria, a group of clinical syndromes attributable to organ dysfunction. Cerebral malaria is an infrequent but life-threatening complication of severe malaria that presents as an acute cerebrovascular encephalopathy characterized by unarousable coma. Despite effective antiparasite drug treatment, 20% of patients with cerebral malaria die from this disease, and many survivors of cerebral malaria have neurocognitive impairment. Thus, an important unmet clinical need is to rapidly identify people with malaria who are at risk of developing cerebral malaria and to develop preventive, adjunctive and neuroprotective treatments for cerebral malaria. This Review describes important advances in the understanding of cerebral malaria over the past two decades and discusses how these mechanistic insights could be translated into new therapies.
Collapse
Affiliation(s)
- Alexandros Hadjilaou
- Protozoen Immunologie, Bernhard-Nocht-Institut für Tropenmedizin (BNITM), Hamburg, Germany.
- Institut für Neuroimmunologie und Multiple Sklerose, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.
| | - Johannes Brandi
- Protozoen Immunologie, Bernhard-Nocht-Institut für Tropenmedizin (BNITM), Hamburg, Germany
| | - Mathias Riehn
- Protozoen Immunologie, Bernhard-Nocht-Institut für Tropenmedizin (BNITM), Hamburg, Germany
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Jacobs
- Protozoen Immunologie, Bernhard-Nocht-Institut für Tropenmedizin (BNITM), Hamburg, Germany
| |
Collapse
|
4
|
Álvarez L, Marín-García PJ, Llobat L. Genetic haplotypes associated with immune response to Leishmania infantum infection in dogs. Vet Res Commun 2023; 47:1675-1685. [PMID: 37059873 DOI: 10.1007/s11259-023-10123-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: 02/17/2023] [Accepted: 04/10/2023] [Indexed: 04/16/2023]
Abstract
Leishmaniasis is a zoonotic parasitic disease, and the main reservoir of the parasite is the dog, although recent years have seen an increase in other mammalian species. In the Mediterranean region, where it is an endemic disease, it is caused by the species Leishmania infantum. The Ibizan hound, an autochthonous breed of this region, appears to have a genetic resistance to parasitic infection, whereas other canine breeds, such as the Boxer, are susceptible to infection. These differences are related to the differentiated activation of the immune response, with the Ibizan hound activating the Th1 immune response, whereas the Boxer breed triggers the Th2 immune response. Cytokine levels and genomic haplotypes of several genes involved in the immune response were analysed in twenty-eight Ibizan hound (resistant canine breed model) and twenty-four Boxer (susceptible canine breed) without clinical signs in the Mediterranean region. Cytokine levels were analysed by ELISA commercial kits and haplotypes were studied using CanineHD DNA Analysis BeadChip including 165,480 mapped positions. The results show 126 haplotypes associated with differential immune response in dogs. Specifically, haplotypes in IL12RB1, IL6R, CIITA, THEMIS, NOXA1, HEY2, RAB38, SLC35D2, SLC28A3, RASEF and DAPK1 genes are associated with serum levels of IFN-γ, IL-2, IL-8, and IL-18. These results suggest that the resistance or susceptibility to Leishmania infantum infection could be a consequence of haplotypes in several genes related to immune response. Future studies are needed to elucidate the relationship of these haplotypes with immune response and gene expression regulation.
Collapse
Affiliation(s)
- Luis Álvarez
- Departamento Produccion Y Sanidad Animal, Salud Publica Y Ciencia Y Tecnologia de los Alimentos, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Pablo-Jesús Marín-García
- Departamento Produccion Y Sanidad Animal, Salud Publica Y Ciencia Y Tecnologia de los Alimentos, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Lola Llobat
- Departamento Produccion Y Sanidad Animal, Salud Publica Y Ciencia Y Tecnologia de los Alimentos, Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain.
| |
Collapse
|
5
|
Azzuolo A, Yang Y, Berghuis A, Fodil N, Gros P. Biphosphoglycerate Mutase: A Novel Therapeutic Target for Malaria? Transfus Med Rev 2023; 37:150748. [PMID: 37827586 DOI: 10.1016/j.tmrv.2023.150748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 10/14/2023]
Abstract
Biphosphoglycerate mutase (BPGM) is a tri-functional enzyme expressed exclusively in erythroid cells and tissues that is responsible for the production of 2,3-biphosphoglycerate (2,3-BPG) through the Rapoport-Luebering shunt. The 2,3-BPG is required for efficient glycolysis and ATP production under anaerobic conditions, but is also a critical allosteric regulator of hemoglobin (Hb), acting to regulate oxygen release in peripheral tissues. In humans, BPGM deficiency is very rare, and is associated with reduced levels of erythrocytic 2,3-BPG and ATP, left shifted Hb-O2 dissociation curve, low P50, elevated Hb and constitutive erythrocytosis. BPGM deficiency in mice recapitulates the erythroid defects seen in human patients. A recent report has shown that BPGM deficiency in mice affords striking protection against both severe malaria anemia and cerebral malaria. These findings are reminiscent of studies of another erythrocyte specific glycolytic enzyme, Pyruvate Kinase (PKLR), which mutational inactivation protects humans and mice against malaria through impairment of glycolysis and ATP production in erythrocytes. BPGM, and PKLR join glucose-6-phosphate dehydrogenase (G6PD) and other erythrocyte variants as modulating response to malaria. Recent studies reviewed suggest glycolysis in general, and BPGM in particular, as a novel pharmacological target for therapeutic intervention in malaria.
Collapse
Affiliation(s)
- Alessia Azzuolo
- Department of Biochemistry, Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada; Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada
| | - Yunxiang Yang
- Department of Biochemistry, Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada; Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada
| | - Albert Berghuis
- Department of Biochemistry, Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada
| | - Nassima Fodil
- Department of Biochemistry, Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada; Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada
| | - Philippe Gros
- Department of Biochemistry, Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada; Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
6
|
Tang J, Jia X, Li J, Dong J, Wang J, Li W, Zhu Y, Hu Y, Hou B, Lin C, Cong Y, Ren T, Yan C, Yang H, Lai Q, Zheng H, Bao Y, Gautam N, Wang HR, Xu B, Chen XL, Li Q, Gascoigne NRJ, Fu G. Themis suppresses the effector function of CD8 + T cells in acute viral infection. Cell Mol Immunol 2023; 20:512-524. [PMID: 36977779 PMCID: PMC10203318 DOI: 10.1038/s41423-023-00997-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
CD8+ T cells play a central role in antiviral immune responses. Upon infection, naive CD8+ T cells differentiate into effector cells to eliminate virus-infected cells, and some of these effector cells further differentiate into memory cells to provide long-term protection after infection is resolved. Although extensively investigated, the underlying mechanisms of CD8+ T-cell differentiation remain incompletely understood. Themis is a T-cell-specific protein that plays critical roles in T-cell development. Recent studies using Themis T-cell conditional knockout mice also demonstrated that Themis is required to promote mature CD8+ T-cell homeostasis, cytokine responsiveness, and antibacterial responses. In this study, we used LCMV Armstrong infection as a probe to explore the role of Themis in viral infection. We found that preexisting CD8+ T-cell homeostasis defects and cytokine hyporesponsiveness do not impair viral clearance in Themis T-cell conditional knockout mice. Further analyses showed that in the primary immune response, Themis deficiency promoted the differentiation of CD8+ effector cells and increased their TNF and IFNγ production. Moreover, Themis deficiency impaired memory precursor cell (MPEC) differentiation but promoted short-lived effector cell (SLEC) differentiation. Themis deficiency also enhanced effector cytokine production in memory CD8+ T cells while impairing central memory CD8+ T-cell formation. Mechanistically, we found that Themis mediates PD-1 expression and its signaling in effector CD8+ T cells, which explains the elevated cytokine production in these cells when Themis is disrupted.
Collapse
Affiliation(s)
- Jian Tang
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Xian Jia
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Jian Li
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Junchen Dong
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Jiayu Wang
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Wanyun Li
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Yuzhen Zhu
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Yanyan Hu
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Bowen Hou
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Chunjie Lin
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Yu Cong
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Tong Ren
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Changsheng Yan
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Hongying Yang
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Qian Lai
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Haiping Zheng
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Yuzhou Bao
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Namrata Gautam
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hong-Rui Wang
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Xiao Lei Chen
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
| | - Qing Li
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
| | - Nicholas R J Gascoigne
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Guo Fu
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
- Department of Hematology, The First Affiliated Hospital and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China.
- Cancer Research Center of Xiamen University, Xiamen, China.
| |
Collapse
|
7
|
Abstract
Immunity to infection has been extensively studied in humans and mice bearing naturally occurring or experimentally introduced germline mutations. Mouse studies are sometimes neglected by human immunologists, on the basis that mice are not humans and the infections studied are experimental and not natural. Conversely, human studies are sometimes neglected by mouse immunologists, on the basis of the uncontrolled conditions of study and small numbers of patients. However, both sides would agree that the infectious phenotypes of patients with inborn errors of immunity often differ from those of the corresponding mutant mice. Why is that? We argue that this important question is best addressed by revisiting and reinterpreting the findings of both mouse and human studies from a genetic perspective. Greater caution is required for reverse-genetics studies than for forward-genetics studies, but genetic analysis is sufficiently strong to define the studies likely to stand the test of time. Genetically robust mouse and human studies can provide invaluable complementary insights into the mechanisms of immunity to infection common and specific to these two species.
Collapse
Affiliation(s)
- Philippe Gros
- McGill University Research Center on Complex Traits, Department of Biochemistry, and Department of Human Genetics, McGill University, Montréal, Québec, Canada;
| | - Jean-Laurent Casanova
- Howard Hughes Medical Institute and St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA;
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, and University of Paris Cité, Imagine Institute and Necker Hospital for Sick Children, Paris, France
| |
Collapse
|
8
|
Exopolyphosphatases PPX1 and PPX2 from Mycobacterium tuberculosis regulate dormancy response and pathogenesis. Microb Pathog 2022; 173:105885. [DOI: 10.1016/j.micpath.2022.105885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/12/2022] [Accepted: 11/12/2022] [Indexed: 11/21/2022]
|
9
|
Sánchez-Arcila JC, Jensen KDC. Forward Genetics in Apicomplexa Biology: The Host Side of the Story. Front Cell Infect Microbiol 2022; 12:878475. [PMID: 35646724 PMCID: PMC9133346 DOI: 10.3389/fcimb.2022.878475] [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: 02/18/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Forward genetic approaches have been widely used in parasitology and have proven their power to reveal the complexities of host-parasite interactions in an unbiased fashion. Many aspects of the parasite's biology, including the identification of virulence factors, replication determinants, antibiotic resistance genes, and other factors required for parasitic life, have been discovered using such strategies. Forward genetic approaches have also been employed to understand host resistance mechanisms to parasitic infection. Here, we will introduce and review all forward genetic approaches that have been used to identify host factors involved with Apicomplexa infections, which include classical genetic screens and QTL mapping, GWAS, ENU mutagenesis, overexpression, RNAi and CRISPR-Cas9 library screens. Collectively, these screens have improved our understanding of host resistance mechanisms, immune regulation, vaccine and drug designs for Apicomplexa parasites. We will also discuss how recent advances in molecular genetics give present opportunities to further explore host-parasite relationships.
Collapse
Affiliation(s)
- Juan C. Sánchez-Arcila
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
| | - Kirk D. C. Jensen
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
- Health Science Research Institute, University of California, Merced, Merced, CA, United States
| |
Collapse
|
10
|
Mélique S, Yang C, Lesourne R. Negative times negative equals positive, THEMIS sets the rule on thymic selection and peripheral T cell responses. Biomed J 2022; 45:334-346. [PMID: 35346866 PMCID: PMC9250082 DOI: 10.1016/j.bj.2022.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/03/2022] [Accepted: 03/03/2022] [Indexed: 12/31/2022] Open
Abstract
The activity of T cells is finely controlled by a set of negative regulators of T-cell antigen receptor (TCR)-mediated signaling. However, how those negative regulators are themselves controlled to prevent ineffective TCR-mediated responses remain poorly understood. Thymocyte-expressed molecule involved in selection (THEMIS) has been characterized over a decade ago as an important player of T cell development. Although the molecular function of THEMIS has long remained puzzling and subject to controversies, latest investigations suggest that THEMIS stimulates TCR-mediated signaling by repressing the tyrosine phosphatases SHP-1 and SHP-2 which exert regulatory function on T cell activation. Recent evidences also point to a role for THEMIS in peripheral T cells beyond its role on thymic selection. Here, we present an overview of the past research on THEMIS in the context of T cell development and peripheral T cell function and discuss the possible implication of THEMIS-based mechanisms on TCR-dependent and independent signaling outcomes.
Collapse
Affiliation(s)
- Suzanne Mélique
- Infinity, University of Toulouse, CNRS5051, INSERM1291, UPS, Toulouse, France
| | - Cui Yang
- Infinity, University of Toulouse, CNRS5051, INSERM1291, UPS, Toulouse, France
| | - Renaud Lesourne
- Infinity, University of Toulouse, CNRS5051, INSERM1291, UPS, Toulouse, France.
| |
Collapse
|
11
|
Liu Y, Cong Y, Niu Y, Yuan Y, Tan F, Lai Q, Hu Y, Hou B, Li J, Lin C, Zheng H, Dong J, Tang J, Chen Q, Brzostek J, Zhang X, Chen XL, Wang HR, Gascoigne NRJ, Xu B, Lin SH, Fu G. Themis is indispensable for IL-2 and IL-15 signaling in T cells. Sci Signal 2022; 15:eabi9983. [PMID: 35167340 DOI: 10.1126/scisignal.abi9983] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
To perform their antiviral and antitumor functions, T cells must integrate signals both from the T cell receptor (TCR), which instruct the cell to remain quiescent or become activated, and from cytokines that guide cellular proliferation and differentiation. In mature CD8+ T cells, Themis has been implicated in integrating TCR and cytokine signals. We investigated whether Themis plays a direct role in cytokine signaling in mature T cells. Themis was required for IL-2- and IL-15-driven CD8+ T cell proliferation both in mice and in vitro. Mechanistically, we found that Themis promoted the activation of the transcription factor Stat and mechanistic target of rapamycin signaling downstream of cytokine receptors. Metabolomics and stable isotope tracing analyses revealed that Themis deficiency reduced glycolysis and serine and nucleotide biosynthesis, demonstrating a receptor-proximal requirement for Themis in triggering the metabolic changes that enable T cell proliferation. The cellular, metabolic, and biochemical defects caused by Themis deficiency were corrected in mice lacking both Themis and the phosphatase Shp1, suggesting that Themis mediates IL-2 and IL-15 receptor-proximal signaling by restraining the activity of Shp1. Together, these results not only shed light on the mechanisms of cytokine signaling but also provide new clues on manipulating T cells for clinical applications.
Collapse
Affiliation(s)
- Yongchao Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yu Cong
- Department of Hematology, First Affiliated Hospital and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China.,Cancer Research Center of Xiamen University, Xiamen, China
| | - Yujia Niu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yin Yuan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Fancheng Tan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Qian Lai
- Department of Hematology, First Affiliated Hospital and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Yanyan Hu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Bowen Hou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Jian Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Chunjie Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Haiping Zheng
- Department of Hematology, First Affiliated Hospital and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Junchen Dong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Jian Tang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Qinwei Chen
- Department of Hematology, First Affiliated Hospital and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Joanna Brzostek
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xueqin Zhang
- Department of Obstetrics and Gynecology, Affiliated Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xiao Lei Chen
- Department of Hematology, First Affiliated Hospital and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Hong-Rui Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China.,Department of Obstetrics and Gynecology, Affiliated Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Nicholas R J Gascoigne
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Bing Xu
- Department of Hematology, First Affiliated Hospital and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China.,Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, China
| | - Shu-Hai Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Guo Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China.,Department of Hematology, First Affiliated Hospital and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China.,Cancer Research Center of Xiamen University, Xiamen, China.,Department of Obstetrics and Gynecology, Affiliated Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
12
|
Xu G, van Bruggen R, Gualtieri CO, Moradin N, Fois A, Vallerand D, De Sa Tavares Russo M, Bassenden A, Lu W, Tam M, Lesage S, Girouard H, Avizonis DZ, Deblois G, Prchal JT, Stevenson M, Berghuis A, Muir T, Rabinowitz J, Vidal SM, Fodil N, Gros P. Bisphosphoglycerate Mutase Deficiency Protects against Cerebral Malaria and Severe Malaria-Induced Anemia. Cell Rep 2020; 32:108170. [PMID: 32966787 DOI: 10.1016/j.celrep.2020.108170] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 07/07/2020] [Accepted: 08/27/2020] [Indexed: 11/30/2022] Open
Abstract
The replication cycle and pathogenesis of the Plasmodium malarial parasite involves rapid expansion in red blood cells (RBCs), and variants of certain RBC-specific proteins protect against malaria in humans. In RBCs, bisphosphoglycerate mutase (BPGM) acts as a key allosteric regulator of hemoglobin/oxyhemoglobin. We demonstrate here that a loss-of-function mutation in the murine Bpgm (BpgmL166P) gene confers protection against both Plasmodium-induced cerebral malaria and blood-stage malaria. The malaria protection seen in BpgmL166P mutant mice is associated with reduced blood parasitemia levels, milder clinical symptoms, and increased survival. The protective effect of BpgmL166P involves a dual mechanism that enhances the host's stress erythroid response to Plasmodium-driven RBC loss and simultaneously alters the intracellular milieu of the RBCs, including increased oxyhemoglobin and reduced energy metabolism, reducing Plasmodium maturation, and replication. Overall, our study highlights the importance of BPGM as a regulator of hemoglobin/oxyhemoglobin in malaria pathogenesis and suggests a new potential malaria therapeutic target.
Collapse
Affiliation(s)
- Guoyue Xu
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; McGill University Research Centre on Complex Traits, McGill University, Montreal, QC H3G 0B1, Canada
| | - Rebekah van Bruggen
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC H3G 0B1, Canada; Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada
| | - Christian O Gualtieri
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC H3G 0B1, Canada; Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada
| | - Neda Moradin
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC H3G 0B1, Canada
| | - Adrien Fois
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC H1T 2M4, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Diane Vallerand
- Université de Montréal, Département de Pharmacologie et Physiologie, Pav Roger-Gaudry, 2900 Édouard-Montpetit, Montréal, QC H3T 1J4, Canada
| | | | - Angelia Bassenden
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada
| | - Wenyun Lu
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Mifong Tam
- Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
| | - Sylvie Lesage
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC H1T 2M4, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Hélène Girouard
- Université de Montréal, Département de Pharmacologie et Physiologie, Pav Roger-Gaudry, 2900 Édouard-Montpetit, Montréal, QC H3T 1J4, Canada
| | - Daina Zofija Avizonis
- Rosalind and Morris Goodman Cancer Research Centre, 1160 Pin Avenue West, Montréal, QC H3A 1A3, Canada
| | - Geneviève Deblois
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Faculté de Pharmacie, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Josef T Prchal
- Division of Hematology, School of Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Mary Stevenson
- Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
| | - Albert Berghuis
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada
| | - Tom Muir
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Joshua Rabinowitz
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Silvia M Vidal
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; McGill University Research Centre on Complex Traits, McGill University, Montreal, QC H3G 0B1, Canada; Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
| | - Nassima Fodil
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC H3G 0B1, Canada; Centre CERMO-FC Pavillon des Sciences Biologiques, 141 Avenue du Président Kennedy, Montréal, QC H2X 3Y7, Canada.
| | - Philippe Gros
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada; McGill University Research Centre on Complex Traits, McGill University, Montreal, QC H3G 0B1, Canada; Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada.
| |
Collapse
|
13
|
Olivier JF, Fodil N, Al Habyan S, Gopal A, Artusa P, Mandl JN, McCaffrey L, Gros P. CCDC88B is required for mobility and inflammatory functions of dendritic cells. J Leukoc Biol 2020; 108:1787-1802. [PMID: 32480428 DOI: 10.1002/jlb.3a0420-386r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 04/13/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022] Open
Abstract
The Coiled Coil Domain Containing Protein 88B (CCDC88B) gene is associated with susceptibility to several inflammatory diseases in humans and its inactivation in mice protects against acute neuroinflammation and models of intestinal colitis. We report that mice lacking functional CCDC88B (Ccdc88bMut ) are defective in several dendritic cells (DCs)-dependent inflammatory and immune reactions in vivo. In these mice, an inflammatory stimulus (LPS) fails to induce the recruitment of DCs into the draining lymph nodes (LNs). In addition, OVA-pulsed Ccdc88bMut DCs injected in the footpad do not induce recruitment and activation of antigen-specific CD4+ and CD8+ T cells in their draining LN. Experiments in vitro indicate that this defect is independent of the ability of mutant DCs to capture and present peptide antigen to T cells. Rather, kinetic analyses in vivo of wild-type and Ccdc88bMut DCs indicate a reduced migration capacity in the absence of the CCDC88B protein expression. Moreover, using time-lapse light microscopy imaging, we show that Ccdc88bMut DCs have an intrinsic motility defect. Furthermore, in vivo studies reveal that these reduced migratory properties lead to dampened contact hypersensitivity reactions in Ccdc88b mutant mice. These findings establish a critical role of CCDC88B in regulating movement and migration of DCs. Thus, regulatory variants impacting Ccdc88b expression in myeloid cells may cause variable degrees of DC-dependent inflammatory response in situ, providing a rationale for the genetic association of CCDC88B with several inflammatory and autoimmune diseases in humans.
Collapse
Affiliation(s)
- Jean-Frederic Olivier
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,McGill Research Center for Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Nassima Fodil
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,McGill Research Center for Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Sara Al Habyan
- Department of Oncology, McGill University, Montreal, Quebec, Canada.,Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada
| | - Angelica Gopal
- McGill Research Center for Complex Traits, McGill University, Montreal, Quebec, Canada.,Department of Chemistry, McGill University, Montreal, Quebec, Canada.,Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Patricio Artusa
- McGill Research Center for Complex Traits, McGill University, Montreal, Quebec, Canada.,Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Judith N Mandl
- McGill Research Center for Complex Traits, McGill University, Montreal, Quebec, Canada.,Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Luke McCaffrey
- Department of Oncology, McGill University, Montreal, Quebec, Canada.,Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada
| | - Philippe Gros
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,McGill Research Center for Complex Traits, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
14
|
ZBTB7B (ThPOK) Is Required for Pathogenesis of Cerebral Malaria and Protection against Pulmonary Tuberculosis. Infect Immun 2020; 88:IAI.00845-19. [PMID: 31792077 DOI: 10.1128/iai.00845-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 11/20/2022] Open
Abstract
We used a genome-wide screen in N-ethyl-N-nitrosourea (ENU)-mutagenized mice to identify genes in which recessive loss-of-function mutations protect against pathological neuroinflammation. We identified an R367Q mutation in the ZBTB7B (ThPOK) protein in which homozygosity causes protection against experimental cerebral malaria (ECM) caused by infection with Plasmodium berghei ANKA. Zbtb7bR367Q homozygous mice show a defect in the lymphoid compartment expressed as severe reduction in the number of single-positive CD4 T cells in the thymus and in the periphery, reduced brain infiltration of proinflammatory leukocytes in P. berghei ANKA-infected mice, and reduced production of proinflammatory cytokines by primary T cells ex vivo and in vivo Dampening of proinflammatory immune responses in Zbtb7bR367Q mice is concomitant to increased susceptibility to infection with avirulent (Mycobacterium bovis BCG) and virulent (Mycobacterium tuberculosis H37Rv) mycobacteria. The R367Q mutation maps to the first DNA-binding zinc finger domain of ThPOK and causes loss of base contact by R367 in the major groove of the DNA, which is predicted to impair DNA binding. Global immunoprecipitation of ThPOK-containing chromatin complexes coupled to DNA sequencing (ChIP-seq) identified transcriptional networks and candidate genes likely to play key roles in CD4+ CD8+ T cell development and in the expression of lineage-specific functions of these cells. This study highlights ThPOK as a global regulator of immune function in which alterations may affect normal responses to infectious and inflammatory stimuli.
Collapse
|
15
|
Jeyakumar T, Beauchemin N, Gros P. Impact of the Microbiome on the Human Genome. Trends Parasitol 2019; 35:809-821. [PMID: 31451407 DOI: 10.1016/j.pt.2019.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 02/07/2023]
Abstract
Humans live in a microbial world that includes pathogenic bacteria, viruses, and fungi that cause lethal infections. In addition, a large number of microbial communities inhabit mucosal surfaces where they provide key metabolic activities, facilitating adaptation to changing environments. New genome technologies enable both sequencing of the human genome and sequence-based cataloging of microbial communities inhabiting human mucosal surfaces. These have revealed intricate two-way relationships between the microbiome and the genome, including strong effects of human genotypes on the composition and activity of the microbiome. Likewise, the microbiome plays an important role in training and regulating the immune system, and acts to modify expression of human genetic risk for debilitating chronic inflammatory and immune conditions. These studies are suggesting a new role of the microbiome in human health and disease.
Collapse
Affiliation(s)
- Thiviya Jeyakumar
- Department of Biochemistry, McGill University, Montreal, Canada; McGill Center for the Study of Complex Traits, McGill University, Montreal, Canada
| | - Nicole Beauchemin
- Department of Biochemistry, McGill University, Montreal, Canada; Goodman Cancer Research Center, McGill University, Montreal, Canada
| | - Philippe Gros
- Department of Biochemistry, McGill University, Montreal, Canada; McGill Center for the Study of Complex Traits, McGill University, Montreal, Canada; Goodman Cancer Research Center, McGill University, Montreal, Canada.
| |
Collapse
|
16
|
Huang HM, McMorran BJ, Foote SJ, Burgio G. Host genetics in malaria: lessons from mouse studies. Mamm Genome 2018; 29:507-522. [PMID: 29594458 DOI: 10.1007/s00335-018-9744-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 03/22/2018] [Indexed: 01/09/2023]
Abstract
Malaria remains a deadly parasitic disease caused by Plasmodium, claiming almost half a million lives every year. While parasite genetics and biology are often the major targets in many studies, it is becoming more evident that host genetics plays a crucial role in the outcome of the infection. Similarly, Plasmodium infections in mice also rely heavily on the genetic background of the mice, and often correlate with observations in human studies, due to their high genetic homology with humans. As such, murine models of malaria are a useful tool for understanding host responses during Plasmodium infections, as well as dissecting host-parasite interactions through various genetic manipulation techniques. Reverse genetic approach such as quantitative trait loci studies and random mutagenesis screens have been employed to discover novel host genes that affect malaria susceptibility in mouse models, while other targeted studies utilize mouse models to validate observation from human studies. Herein, we review the findings from the past and present studies on murine models of hepatic and erythrocytic stages of malaria and speculate on how the current mouse models benefit from the recent development in CRISPR/Cas9 gene editing technology.
Collapse
Affiliation(s)
- Hong Ming Huang
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, 131 Garran Road, Canberra, ACT, 2601, Australia
| | - Brendan J McMorran
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, 131 Garran Road, Canberra, ACT, 2601, Australia
| | - Simon J Foote
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, 131 Garran Road, Canberra, ACT, 2601, Australia
| | - Gaetan Burgio
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, 131 Garran Road, Canberra, ACT, 2601, Australia.
| |
Collapse
|
17
|
Genetic analysis of cerebral malaria in the mouse model infected with Plasmodium berghei. Mamm Genome 2018; 29:488-506. [DOI: 10.1007/s00335-018-9752-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 06/05/2018] [Indexed: 12/22/2022]
|
18
|
Rocaglates as dual-targeting agents for experimental cerebral malaria. Proc Natl Acad Sci U S A 2018; 115:E2366-E2375. [PMID: 29463745 DOI: 10.1073/pnas.1713000115] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cerebral malaria (CM) is a severe and rapidly progressing complication of infection by Plasmodium parasites that is associated with high rates of mortality and morbidity. Treatment options are currently few, and intervention with artemisinin (Art) has limited efficacy, a problem that is compounded by the emergence of resistance to Art in Plasmodium parasites. Rocaglates are a class of natural products derived from plants of the Aglaia genus that have been shown to interfere with eukaryotic initiation factor 4A (eIF4A), ultimately blocking initiation of protein synthesis. Here, we show that the rocaglate CR-1-31B perturbs association of Plasmodium falciparum eIF4A (PfeIF4A) with RNA. CR-1-31B shows potent prophylactic and therapeutic antiplasmodial activity in vivo in mouse models of infection with Plasmodium berghei (CM) and Plasmodium chabaudi (blood-stage malaria), and can also block replication of different clinical isolates of P. falciparum in human erythrocytes infected ex vivo, including drug-resistant P. falciparum isolates. In vivo, a single dosing of CR-1-31B in P. berghei-infected animals is sufficient to provide protection against lethality. CR-1-31B is shown to dampen expression of the early proinflammatory response in myeloid cells in vitro and dampens the inflammatory response in vivo in P. berghei-infected mice. The dual activity of CR-1-31B as an antiplasmodial and as an inhibitor of the inflammatory response in myeloid cells should prove extremely valuable for therapeutic intervention in human cases of CM.
Collapse
|
19
|
Finding the 'ubiquitous' threads in infection and autoimmune neuroinflammation. Nat Immunol 2017; 18:7-8. [PMID: 27984564 DOI: 10.1038/ni.3633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
20
|
USP15 regulates type I interferon response and is required for pathogenesis of neuroinflammation. Nat Immunol 2016; 18:54-63. [DOI: 10.1038/ni.3581] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 08/25/2016] [Indexed: 12/21/2022]
|
21
|
Moradin N, Torre S, Gauthier S, Tam M, Hawari J, Vandercruyssen K, De Spiegeleer B, Fortin A, Stevenson MM, Gros P. Cysteamine broadly improves the anti-plasmodial activity of artemisinins against murine blood stage and cerebral malaria. Malar J 2016; 15:260. [PMID: 27150250 PMCID: PMC4858922 DOI: 10.1186/s12936-016-1317-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 04/28/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The potential emergence and spread of resistance to artemisinins in the Plasmodium falciparum malaria parasite constitutes a major global health threat. Hence, improving the efficacy of artemisinins and of artemisinin-based combination therapy (ACT) represents a major short-term goal in the global fight against malaria. Mice defective in the enzyme pantetheinase (Vnn3) show increased susceptibility to blood-stage malaria (increased parasitaemia, reduced survival), and supplementation of Vnn3 mutants with the reaction product of pantetheinase, cysteamine, corrects in part the malaria-susceptibility phenotype of the mutants. Cysteamine (Cys) is a small, naturally occurring amino-thiol that has very low toxicity in vivo and is approved for clinical use in the life-long treatment of the kidney disorder nephropathic cystinosis. METHODS The ability of Cys to improve the anti-plasmodial activity of different clinically used artemisinins was tested. The effect of different CYS/ART combinations on malarial phenotypes (parasite blood-stage replication, overall and survival from lethal infection) was assessed in a series of in vivo experiments using Plasmodium strains that induce either blood-stage (Plasmodium chabaudi AS) or cerebral disease (Plasmodium berghei ANKA). This was also evaluated in an ex vivo experimental protocol that directly assesses the effect of such drug combinations on the viability of Plasmodium parasites, as measured by the ability of tested parasites to induce a productive infection in vivo in otherwise naïve animals. RESULTS Cys is found to potentiate the anti-plasmodial activity of artesunate, artemether, and arteether, towards the blood-stage malaria parasite P. chabaudi AS. Ex vivo experiments, indicate that potentiation of the anti-plasmodial activity of artemisinins by Cys is direct and does not require the presence of host factors. In addition, potentiation occurs at sub-optimal concentrations of artemisinins and Cys that on their own have little or no effect on parasite growth. Cys also dramatically enhances the efficacy and protective effect of artemisinins against cerebral malaria induced by infection with the P. berghei ANKA parasite. CONCLUSION These findings indicate that inclusion of Cys in current formulations of ACT, or its use as adjunct therapy could improve the anti-plasmodial activity of artemisinin, decrease mortality in cerebral malaria patients, and prevent or delay the development and spread of artemisinin resistance.
Collapse
Affiliation(s)
- Neda Moradin
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, room 366, Montreal, QC, H3G 0B1, Canada
| | - Sabrina Torre
- Department of Human Genetics, McGill University, Montreal, Canada
| | - Susan Gauthier
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, room 366, Montreal, QC, H3G 0B1, Canada
| | - Mifong Tam
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Jalal Hawari
- Department of Civil, Geological and Mining Engineering, Ecole Polytechnique, Université de Montreal, Montreal, Canada
| | | | | | - Anny Fortin
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, room 366, Montreal, QC, H3G 0B1, Canada
| | - Mary M Stevenson
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Philippe Gros
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, room 366, Montreal, QC, H3G 0B1, Canada.
| |
Collapse
|
22
|
A Point Mutation in p190A RhoGAP Affects Ciliogenesis and Leads to Glomerulocystic Kidney Defects. PLoS Genet 2016; 12:e1005785. [PMID: 26859289 PMCID: PMC4747337 DOI: 10.1371/journal.pgen.1005785] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 12/12/2015] [Indexed: 01/09/2023] Open
Abstract
Rho family GTPases act as molecular switches regulating actin cytoskeleton dynamics. Attenuation of their signaling capacity is provided by GTPase-activating proteins (GAPs), including p190A, that promote the intrinsic GTPase activity of Rho proteins. In the current study we have performed a small-scale ENU mutagenesis screen and identified a novel loss of function allele of the p190A gene Arhgap35, which introduces a Leu1396 to Gln substitution in the GAP domain. This results in decreased GAP activity for the prototypical Rho-family members, RhoA and Rac1, likely due to disrupted ordering of the Rho binding surface. Consequently, Arhgap35-deficient animals exhibit hypoplastic and glomerulocystic kidneys. Investigation into the cystic phenotype shows that p190A is required for appropriate primary cilium formation in renal nephrons. P190A specifically localizes to the base of the cilia to permit axoneme elongation, which requires a functional GAP domain. Pharmacological manipulations further reveal that inhibition of either Rho kinase (ROCK) or F-actin polymerization is able to rescue the ciliogenesis defects observed upon loss of p190A activity. We propose a model in which p190A acts as a modulator of Rho GTPases in a localized area around the cilia to permit the dynamic actin rearrangement required for cilia elongation. Together, our results establish an unexpected link between Rho GTPase regulation, ciliogenesis and glomerulocystic kidney disease.
Collapse
|
23
|
Gascoigne NRJ, Acuto O. THEMIS: a critical TCR signal regulator for ligand discrimination. Curr Opin Immunol 2015; 33:86-92. [PMID: 25700024 DOI: 10.1016/j.coi.2015.01.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 01/27/2015] [Accepted: 01/30/2015] [Indexed: 10/24/2022]
Abstract
Genetic approaches identified THEMIS as a critical element driving positive selection of CD4(+)CD8(+) thymocytes towards maturation. THEMIS is expressed only in the T-cell lineage, and is recruited to the proximity of signaling T-cell antigen receptors (TCR) by association with the membrane scaffold LAT. However, its molecular role remained an enigma until recently. Conventionally positively-selected T-cells are lacking in THEMIS-deficient mice, leading to the initial hypothesis that THEMIS positively regulates TCR signaling. Recent data show that THEMIS deficiency increases rather than decreases TCR signaling, leading to augmented apoptosis. The finding that THEMIS is constitutively bound to the tyrosine phosphatases SHP1 or SHP2, provides a mechanism for THEMIS action. When recruited onto LAT, THEMIS-SHP promotes immediate dephosphorylation of TCR-proximal signaling components. This negative feedback is central in setting sharp signaling thresholds and helps explain the exquisite ligand discrimination by the TCR, particularly during thymocyte selection.
Collapse
Affiliation(s)
- Nicholas R J Gascoigne
- Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 5 Science Drive 2, Singapore 117597, Singapore.
| | - Oreste Acuto
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK.
| |
Collapse
|