1
|
Nath P, Maiti D. A review of the mutagenic potential of N-ethyl-N-nitrosourea (ENU) to induce hematological malignancies. J Biochem Mol Toxicol 2022; 36:e23067. [PMID: 35393684 DOI: 10.1002/jbt.23067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 11/05/2021] [Accepted: 03/23/2022] [Indexed: 12/12/2022]
Abstract
This review is intended to summarize the existing literature on the mutagenicity of N-ethyl-N-nitrosourea (ENU) in inducing hematological malignancies, including acute myeloid leukemia (AML) in mice. Blood or hematological malignancies are the most common malignant disorders seen in people of all age groups. Driven by a number of genetic alterations, leukemia rule out the normal proliferation and differentiation of hematopoietic stem cells (HSCs) and their progenitors in the bone marrow (BM) and severely affects blood functions. Out of all hematological malignancies, AML is the most aggressive type, with a high incidence and mortality rate. AML is found as either de novo or secondary therapeutic AML (t-AML). t-AML is a serious adverse consequence of alkylator chemotherapy to the cancer patient and alone constitutes about 10%-20% of all reported AML cases. Cancer patients who received alkylator chemotherapy are at an elevated risk of developing t-AML. ENU has a long history of use as a potent carcinogen that induces blood malignancies in mice and rats that are pathologically similar to human AML and t-AML. ENU, once entered into the body, circulates all over the body tissues and reaches BM. It creates an overall state of suppression within the BM by damaging the marrow cells, alkylating the DNA, and forming DNA adducts within the early and late hematopoietic stem and progenitor cells. The BM holds a weak DNA repair mechanism due to low alkyltransferase, and poly [ADP-ribose] polymerase (PARP) enzyme content often fails to obliterate those adducts, acting as a catalyst to bring genetic abnormalities, including point gene mutations as well as chromosomal alterations, for example, translocation and inversion. Taking advantage of ENU-induced immune-suppressed state and weak immune surveillance, these mutations remain viable and slowly give rise to transformed HSCs. This review also highlights the carcinogenic nature of ENU and the complex relation between the ENU's overall toxicity in the induction of hematological malignancies.
Collapse
Affiliation(s)
- Priyatosh Nath
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Agartala, Tripura, India
| | - Debasish Maiti
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Agartala, Tripura, India
| |
Collapse
|
2
|
Bourgeois JS, Smith CM, Ko DC. These Are the Genes You're Looking For: Finding Host Resistance Genes. Trends Microbiol 2021; 29:346-362. [PMID: 33004258 PMCID: PMC7969353 DOI: 10.1016/j.tim.2020.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/21/2022]
Abstract
Humanity's ongoing struggle with new, re-emerging and endemic infectious diseases serves as a frequent reminder of the need to understand host-pathogen interactions. Recent advances in genomics have dramatically advanced our understanding of how genetics contributes to host resistance or susceptibility to bacterial infection. Here we discuss current trends in defining host-bacterial interactions at the genome-wide level, including screens that harness CRISPR/Cas9 genome editing, natural genetic variation, proteomics, and transcriptomics. We report on the merits, limitations, and findings of these innovative screens and discuss their complementary nature. Finally, we speculate on future innovation as we continue to progress through the postgenomic era and towards deeper mechanistic insight and clinical applications.
Collapse
Affiliation(s)
- Jeffrey S Bourgeois
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Clare M Smith
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, School of Medicine, Duke University Durham, NC, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA; Division of Infectious Diseases, Department of Medicine, School of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
3
|
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: 1.0] [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
|
4
|
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: 4] [Impact Index Per Article: 1.0] [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
|
5
|
Chehboun S, Leiva-Torres GA, Charbonneau B, Eveleigh R, Bourque G, Vidal SM. A point mutation in the linker domain of mouse STAT5A is associated with impaired NK-cell regulation. Genes Immun 2019; 21:136-141. [PMID: 31591503 DOI: 10.1038/s41435-019-0088-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 08/08/2019] [Accepted: 08/14/2019] [Indexed: 12/12/2022]
Abstract
The transcription factor STAT5 is critical for peripheral NK-cell survival, proliferation, and cytotoxic function. STAT5 refers to two highly related proteins, STAT5A and STAT5B. In this study, we verified the importance of STAT5A isoform for NK cells. We characterized an incidental chemically induced W484G mutation in the Stat5a gene and found that this mutation was associated with a reduction of STAT5A protein expression. Closer examination of NK-cell subsets from Stat5a mutant mice showed marked reductions in NK-cell number and maturation. IL-15 treatment of Stat5a mutant NK cells exhibited defective induction of both STAT5 and mTOR signaling pathways and reduced expression of granzyme B and IFN-γ. Finally, we observed that Stat5a mutant mice revealed more tumor growth upon injection of RMA-S tumor cell line. Overall, our results demonstrate that the W484G mutation in the linker domain of STAT5A is sufficient to compromise STAT5A function in NK-cell homeostasis, responsiveness, and tumoricidal function.
Collapse
Affiliation(s)
- Salma Chehboun
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada.,McGill Research Centre on Complex Traits, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Gabriel André Leiva-Torres
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada.,McGill Research Centre on Complex Traits, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Benoît Charbonneau
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada.,McGill Research Centre on Complex Traits, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Robert Eveleigh
- McGill University and Genome Québec Innovation Centre, Montreal, QC, H3A 0G1, Canada
| | - Guillaume Bourque
- McGill University and Genome Québec Innovation Centre, Montreal, QC, H3A 0G1, Canada
| | - Silvia Marina Vidal
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada. .,McGill Research Centre on Complex Traits, McGill University, Montreal, QC, H3G 0B1, Canada.
| |
Collapse
|
6
|
Expression of CD300lf by microglia contributes to resistance to cerebral malaria by impeding the neuroinflammation. Genes Immun 2019; 21:45-62. [PMID: 31501529 DOI: 10.1038/s41435-019-0085-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/30/2019] [Accepted: 06/06/2019] [Indexed: 01/28/2023]
Abstract
Genetic mapping and genome-wide studies provide evidence for the association of several genetic polymorphisms with malaria, a complex pathological disease with multiple severity degrees. We have previously described Berr1and Berr2 as candidate genes identified in the WLA/Pas inbreed mouse strain predisposing to resistance to cerebral malaria (CM) induced by P. berghei ANKA. We report in this study the phenotypic and functional characteristics of a congenic strain we have derived for Berr2WLA allele on the C57BL/6JR (B6) background. B6.WLA-Berr2 was found highly resistant to CM compared to C57BL/6JR susceptible mice. The mechanisms associated with CM resistance were analyzed by combining genotype, transcriptomic and immune response studies. We found that B6.WLA-Berr2 mice showed a reduced parasite sequestration and blood-brain barrier disruption with low CXCR3+ T cell infiltration in the brain along with altered glial cell response upon P. berghei ANKA infection compared to B6. In addition, we have identified the CD300f, belonging to a family of Ig-like encoding genes, as a potential candidate associated with CM resistance. Microglia cells isolated from the brain of infected B6.WLA-Berr2 mice significantly expressed higher level of CD300f compared to CMS mice and were associated with inhibition of inflammatory response.
Collapse
|
7
|
Yamamoto PK, Souza TA, Antiorio ATFB, Zanatto DA, Garcia‐Gomes MDSA, Alexandre‐Ribeiro SR, Oliveira NDS, Menck CFM, Bernardi MM, Massironi SMG, Mori CMC. Genetic and behavioral characterization of a
Kmt2d
mouse mutant, a new model for Kabuki Syndrome. GENES BRAIN AND BEHAVIOR 2019; 18:e12568. [DOI: 10.1111/gbb.12568] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/18/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Pedro K. Yamamoto
- Department of Pathology, School of Veterinary Medicine and Animal ScienceUniversity of São Paulo (USP) Sao Paulo Brazil
| | - Tiago A. Souza
- Department of Microbiology, Institute of Biomedical ScienceUniversity of São Paulo (USP) Sao Paulo Brazil
| | - Ana T. F. B. Antiorio
- Department of Pathology, School of Veterinary Medicine and Animal ScienceUniversity of São Paulo (USP) Sao Paulo Brazil
| | - Dennis A. Zanatto
- Department of Pathology, School of Veterinary Medicine and Animal ScienceUniversity of São Paulo (USP) Sao Paulo Brazil
| | | | | | - Nicassia de Souza Oliveira
- Department of Pathology, School of Veterinary Medicine and Animal ScienceUniversity of São Paulo (USP) Sao Paulo Brazil
| | - Carlos F. M. Menck
- Department of Microbiology, Institute of Biomedical ScienceUniversity of São Paulo (USP) Sao Paulo Brazil
| | - Maria M. Bernardi
- Graduate Program in Environmental and Experimental Pathology, Paulista University São Paulo Brazil
| | - Silvia M. G. Massironi
- Department of Pathology, School of Veterinary Medicine and Animal ScienceUniversity of São Paulo (USP) Sao Paulo Brazil
- Department of Immunology, Institute of Biomedical ScienceUniversity of São Paulo (USP) Sao Paulo Brazil
| | - Claudia M. C. Mori
- Department of Pathology, School of Veterinary Medicine and Animal ScienceUniversity of São Paulo (USP) Sao Paulo Brazil
| |
Collapse
|
8
|
Zhang W, Jia P, Liu W, Jia K, Yi M. Screening for Antiviral Medaka Haploid Embryonic Stem Cells by Genome Wide Mutagenesis. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2019; 21:186-195. [PMID: 30617926 DOI: 10.1007/s10126-018-09870-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/21/2018] [Indexed: 06/09/2023]
Abstract
Nervous necrosis virus (NNV), one of the most prevalent fish pathogens, has caused significant losses in both yield and economy to the aquaculture. Host factors involved in NNV infection remain to be identified due to the lack of ideal model for the study of NNV and host interaction. Haploid stem cells have proven to be ideal materials in genetic screens. Here, we generated a cell line HX1G1 (simply named G1) with the activity against red-spotted grouper nervous necrosis virus (RGNNV) by N-ethyl-N-nitrosourea (ENU)-mediated whole genome random mutagenesis from the haploid embryonic stem cell HX1a, a cell clone from haploid cell line HX1 that we previously derived from the medaka fish. G1 cells retained the characteristics of haploidy and pluripotency as indicated by the EBs differentiation ability after genetic mutagenesis. Compared with HX1a cells, no typical cytopathic effects were observed, and the expression of RNA-dependent RNA polymerase (RDRP) was significantly reduced in G1 cells post RGNNV infection, indicating the enhanced anti-RGNNV activity of G1. Furthermore, we demonstrated that RGNNV entry into G1 cells was partially inhibited, and this inhibition might be relevant to the induced mutation of heat shock cognate protein 70 (HSC70) which was decisive for NNV entry. Interestingly, G1 cells were to some extent permissive to RGNNV infection, but RGNNV was spontaneously cleared in G1 cells during serial passage. In addition, we also found that the expression levels of interferon (IFN)-related genes were higher in G1 cells than those in HX1a cells, suggesting that viral clearance might be associated with the elevated expression of IFN-related genes in G1 cells.
Collapse
Affiliation(s)
- Wanwan Zhang
- School of Marine Sciences, Sun Yat-sen University, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou, China
- Zhuhai Key Laboratory of Marine Bioresources and Environment, Guangdong, China
| | - Peng Jia
- School of Marine Sciences, Sun Yat-sen University, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou, China
- Zhuhai Key Laboratory of Marine Bioresources and Environment, Guangdong, China
| | - Wei Liu
- School of Marine Sciences, Sun Yat-sen University, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou, China
- Zhuhai Key Laboratory of Marine Bioresources and Environment, Guangdong, China
| | - Kuntong Jia
- School of Marine Sciences, Sun Yat-sen University, Guangdong, China.
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou, China.
- Zhuhai Key Laboratory of Marine Bioresources and Environment, Guangdong, China.
| | - Meisheng Yi
- School of Marine Sciences, Sun Yat-sen University, Guangdong, China.
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou, China.
- Zhuhai Key Laboratory of Marine Bioresources and Environment, Guangdong, China.
| |
Collapse
|
9
|
Mancini M, Caignard G, Charbonneau B, Dumaine A, Wu N, Leiva-Torres GA, Gerondakis S, Pearson A, Qureshi ST, Sladek R, Vidal SM. Rel-Dependent Immune and Central Nervous System Mechanisms Control Viral Replication and Inflammation during Mouse Herpes Simplex Encephalitis. THE JOURNAL OF IMMUNOLOGY 2019; 202:1479-1493. [DOI: 10.4049/jimmunol.1800063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 12/21/2018] [Indexed: 01/01/2023]
|
10
|
Genetic approach towards a vaccine against malaria. Eur J Clin Microbiol Infect Dis 2018; 37:1829-1839. [PMID: 29956023 DOI: 10.1007/s10096-018-3313-8] [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: 06/05/2018] [Accepted: 06/20/2018] [Indexed: 10/28/2022]
Abstract
Malaria is a major concern for international health authorities. Millions of people contract it every year in the world due to a parasite of the Plasmodium genus. Due to the complexity of the parasite biology and genetics, there is currently no vaccine against the disease. However, due to the great resistance both to the medicines and to the insecticides used to combat the disease, it has become essential to obtain a vaccine as the necessary tool to prevent transmission and eliminate the disease. The bibliometric data indicate that interest in vaccines has been growing steadily since the 1980s. But nowadays, a powerful tool is used: the Plasmodium genome. This allows us to improve the fight against the disease. Knowing the sequences of the genes that favor the appearance of drug resistance, or those that encode for proteins with greater antigenic response, is a tool that can become fundamental. This article reviews the state of the art on vaccines and genetics, in the fight against malaria, and analyzes the fixed photo that the worldwide research on the disease poses.
Collapse
|
11
|
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: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 06/05/2018] [Indexed: 12/22/2022]
|
12
|
Kang E, Crouse A, Chevallier L, Pontier SM, Alzahrani A, Silué N, Campbell-Valois FX, Montagutelli X, Gruenheid S, Malo D. Enterobacteria and host resistance to infection. Mamm Genome 2018; 29:558-576. [PMID: 29785663 DOI: 10.1007/s00335-018-9749-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023]
Abstract
Enterobacteriaceae are a large family of Gram-negative, non-spore-forming bacteria. Although many species exist as part of the natural flora of animals including humans, some members are associated with both intestinal and extraintestinal diseases. In this review, we focus on members of this family that have important roles in human disease: Salmonella, Escherichia, Shigella, and Yersinia, providing a brief overview of the disease caused by these bacteria, highlighting the contribution of animal models to our understanding of their pathogenesis and of host genetic determinants involved in susceptibility or resistance to infection.
Collapse
Affiliation(s)
- Eugene Kang
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- McGill Research Center on Complex Traits, McGill University, Montreal, QC, Canada
| | - Alanna Crouse
- McGill Research Center on Complex Traits, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Lucie Chevallier
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, École Nationale Vétérinaire d'Alfort, UPEC, Maisons-Alfort, France
- Mouse Genetics Laboratory, Department of Genomes and Genetics, Institut Pasteur, Paris, France
| | - Stéphanie M Pontier
- Department of Chemistry and Biomolecular Sciences, Centre for Chemical and Synthetic Biology, University of Ottawa, Ottawa, ON, Canada
| | - Ashwag Alzahrani
- Department of Chemistry and Biomolecular Sciences, Centre for Chemical and Synthetic Biology, University of Ottawa, Ottawa, ON, Canada
| | - Navoun Silué
- Department of Chemistry and Biomolecular Sciences, Centre for Chemical and Synthetic Biology, University of Ottawa, Ottawa, ON, Canada
| | - François-Xavier Campbell-Valois
- Department of Chemistry and Biomolecular Sciences, Centre for Chemical and Synthetic Biology, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Xavier Montagutelli
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, École Nationale Vétérinaire d'Alfort, UPEC, Maisons-Alfort, France
| | - Samantha Gruenheid
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- McGill Research Center on Complex Traits, McGill University, Montreal, QC, Canada
| | - Danielle Malo
- McGill Research Center on Complex Traits, McGill University, Montreal, QC, Canada.
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
- Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
13
|
Sharma Y, Bashir S, Bhardwaj P, Ahmad A, Khan F. Protein tyrosine phosphatase SHP-1: resurgence as new drug target for human autoimmune disorders. Immunol Res 2017; 64:804-19. [PMID: 27216862 DOI: 10.1007/s12026-016-8805-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Recognition of self-antigen and its destruction by the immune system is the hallmark of autoimmune diseases. During the developmental stages, immune cells are introduced to the self-antigen, for which tolerance develops. The inflammatory insults that break the immune tolerance provoke immune system against self-antigen, progressively leading to autoimmune diseases. SH2 domain containing protein tyrosine phosphatase (PTP), SHP-1, was identified as hematopoietic cell-specific PTP that regulates immune function from developing immune tolerance to mediating cell signaling post-immunoreceptor activation. The extensive research on SHP-1-deficient mice elucidated the diversified role of SHP-1 in immune regulation, and inflammatory process and related disorders such as cancer, autoimmunity, and neurodegenerative diseases. The present review focalizes upon the implication of SHP-1 in the pathogenesis of autoimmune disorders, such as allergic asthma, neutrophilic dermatosis, atopic dermatitis, rheumatoid arthritis, and multiple sclerosis, so as to lay the background in pursuance of developing therapeutic strategies targeting SHP-1. Also, new SHP-1 molecular targets have been suggested like SIRP-α, PIPKIγ, and RIP-1 that may prove to be the focal point for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Yadhu Sharma
- Department of Biochemistry, Faculty of Science, Jamia Hamdard, New Delhi, 110062, India
| | - Samina Bashir
- Department of Biochemistry, Faculty of Science, Jamia Hamdard, New Delhi, 110062, India
| | - Puja Bhardwaj
- Department of Biochemistry, Faculty of Science, Jamia Hamdard, New Delhi, 110062, India
| | - Altaf Ahmad
- Department of Botany, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India
| | - Farah Khan
- Department of Biochemistry, Faculty of Science, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
14
|
An siRNA-based screen in C2C12 myoblasts identifies novel genes involved in myogenic differentiation. Exp Cell Res 2017; 359:145-153. [DOI: 10.1016/j.yexcr.2017.07.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 07/16/2017] [Accepted: 07/31/2017] [Indexed: 11/19/2022]
|
15
|
Habas K, Brinkworth MH, Anderson D. In vitro responses to known in vivo genotoxic agents in mouse germ cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2017; 58:99-107. [PMID: 28205273 DOI: 10.1002/em.22075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 01/29/2017] [Indexed: 06/06/2023]
Abstract
Genotoxic compounds have induced DNA damage in male germ cells and have been associated with adverse clinical outcomes including enhanced risks for maternal, paternal and offspring health. DNA strand breaks represent a great threat to the genomic integrity of germ cells. Such integrity is essential to maintain spermatogenesis and prevent reproduction failure. The Comet assay results revealed that the incubation of isolated germ cells with n-ethyl-n-nitrosourea (ENU), 6-mercaptopurine (6-MP) and methyl methanesulphonate (MMS) led to increase in length of Olive tail moment and % tail DNA when compared with the untreated control cells and these effects were concentration-dependent. All compounds were significantly genotoxic in cultured germ cells. Exposure of isolated germ cells to ENU produced the highest concentration-related increase in both DNA damage and gene expression changes in spermatogonia. Spermatocytes were most sensitive to 6-MP, with DNA damage and gene expression changes while spermatids were particularly susceptible to MMS. Real-time PCR results showed that the mRNA level expression of p53 increased and bcl-2 decreased significantly with the increasing ENU, 6-MP and MMS concentrations in spermatogonia, spermatocytes and spermatids respectively for 24 hr. Both are gene targets for DNA damage response and apoptosis. These observations may help explain the cell alterations caused by ENU, 6-MP and MMS in spermatogonia, spermatocytes and spermatids. Taken together, ENU, 6-MP and MMS induced DNA damage and decreased apoptosis associated gene expression in the germ cells in vitro. Environ. Mol. Mutagen. 58:99-107, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Khaled Habas
- School of Medical Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Martin H Brinkworth
- School of Medical Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Diana Anderson
- School of Medical Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| |
Collapse
|
16
|
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: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 08/25/2016] [Indexed: 12/21/2022]
|
17
|
Eppig JT, Richardson JE, Kadin JA, Ringwald M, Blake JA, Bult CJ. Mouse Genome Informatics (MGI): reflecting on 25 years. Mamm Genome 2015; 26:272-84. [PMID: 26238262 PMCID: PMC4534491 DOI: 10.1007/s00335-015-9589-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 12/02/2022]
Abstract
From its inception in 1989, the mission of the Mouse Genome Informatics (MGI) resource remains to integrate genetic, genomic, and biological data about the laboratory mouse to facilitate the study of human health and disease. This mission is ever more feasible as the revolution in genetics knowledge, the ability to sequence genomes, and the ability to specifically manipulate mammalian genomes are now at our fingertips. Through major paradigm shifts in biological research and computer technologies, MGI has adapted and evolved to become an integral part of the larger global bioinformatics infrastructure and honed its ability to provide authoritative reference datasets used and incorporated by many other established bioinformatics resources. Here, we review some of the major changes in research approaches over that last quarter century, how these changes are reflected in the MGI resource you use today, and what may be around the next corner.
Collapse
Affiliation(s)
- Janan T. Eppig
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME 04609 USA
| | - Joel E. Richardson
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME 04609 USA
| | - James A. Kadin
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME 04609 USA
| | - Martin Ringwald
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME 04609 USA
| | - Judith A. Blake
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME 04609 USA
| | - Carol J. Bult
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME 04609 USA
| |
Collapse
|