Pub Date : 2021-01-01DOI: 10.1016/j.crimmu.2021.06.002
Matthias H. Enders , Ganchimeg Bayarsaikhan , Sonia Ghilas , Yu Cheng Chua , Rose May , Maria N. de Menezes , Zhengyu Ge , Peck Szee Tan , Anton Cozijnsen , Vanessa Mollard , Katsuyuki Yui , Geoffrey I. McFadden , Mireille H. Lahoud , Irina Caminschi , Anthony W. Purcell , Ralf B. Schittenhelm , Lynette Beattie , William R. Heath , Daniel Fernandez-Ruiz
Thorough understanding of the role of CD4 T cells in immunity can be greatly assisted by the study of responses to defined specificities. This requires knowledge of Plasmodium-derived immunogenic epitopes, of which only a few have been identified, especially for the mouse C57BL/6 background. We recently developed a TCR transgenic mouse line, termed PbT-II, that produces CD4+ T cells specific for an MHC class II (I-Ab)-restricted Plasmodium epitope and is responsive to both sporozoites and blood-stage P. berghei. Here, we identify a peptide within the P. berghei heat shock protein 90 as the cognate epitope recognised by PbT-II cells. We show that C57BL/6 mice infected with P. berghei blood-stage induce an endogenous CD4 T cell response specific for this epitope, indicating cells of similar specificity to PbT-II cells are present in the naïve repertoire. Adoptive transfer of in vitro activated TH1-, or particularly TH2-polarised PbT-II cells improved control of P. berghei parasitemia in C57BL/6 mice and drastically reduced the onset of experimental cerebral malaria. Our results identify a versatile, potentially protective MHC-II restricted epitope useful for exploration of CD4 T cell-mediated immunity and vaccination strategies against malaria.
{"title":"Plasmodium berghei Hsp90 contains a natural immunogenic I-Ab-restricted antigen common to rodent and human Plasmodium species","authors":"Matthias H. Enders , Ganchimeg Bayarsaikhan , Sonia Ghilas , Yu Cheng Chua , Rose May , Maria N. de Menezes , Zhengyu Ge , Peck Szee Tan , Anton Cozijnsen , Vanessa Mollard , Katsuyuki Yui , Geoffrey I. McFadden , Mireille H. Lahoud , Irina Caminschi , Anthony W. Purcell , Ralf B. Schittenhelm , Lynette Beattie , William R. Heath , Daniel Fernandez-Ruiz","doi":"10.1016/j.crimmu.2021.06.002","DOIUrl":"10.1016/j.crimmu.2021.06.002","url":null,"abstract":"<div><p>Thorough understanding of the role of CD4 T cells in immunity can be greatly assisted by the study of responses to defined specificities. This requires knowledge of <em>Plasmodium</em>-derived immunogenic epitopes, of which only a few have been identified, especially for the mouse C57BL/6 background. We recently developed a TCR transgenic mouse line, termed PbT-II, that produces CD4<sup>+</sup> T cells specific for an MHC class II (I-A<sup>b</sup>)-restricted <em>Plasmodium</em> epitope and is responsive to both sporozoites and blood-stage <em>P. berghei</em>. Here, we identify a peptide within the <em>P. berghei</em> heat shock protein 90 as the cognate epitope recognised by PbT-II cells. We show that C57BL/6 mice infected with <em>P. berghei</em> blood-stage induce an endogenous CD4 T cell response specific for this epitope, indicating cells of similar specificity to PbT-II cells are present in the naïve repertoire. Adoptive transfer of <em>in vitro</em> activated T<sub>H</sub>1-, or particularly T<sub>H</sub>2-polarised PbT-II cells improved control of <em>P. berghei</em> parasitemia in C57BL/6 mice and drastically reduced the onset of experimental cerebral malaria. Our results identify a versatile, potentially protective MHC-II restricted epitope useful for exploration of CD4 T cell-mediated immunity and vaccination strategies against malaria.</p></div>","PeriodicalId":72750,"journal":{"name":"Current research in immunology","volume":"2 ","pages":"Pages 79-92"},"PeriodicalIF":0.0,"publicationDate":"2021-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.crimmu.2021.06.002","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49128943","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-01-01DOI: 10.1016/j.crimmu.2021.03.001
Marcelo Pires Amaral , Fernanda Caroline Coirada , Juliana de Souza Apostolico , Nádia Tomita , Edgar Ruz Fernandes , Higo Fernando Santos Souza , Rosa Maria Chura-Chambi , Ligia Morganti , Silvia Beatriz Boscardin , Daniela Santoro Rosa
Chikungunya virus (CHIKV) is an arbovirus transmitted to humans mainly by the bite of infected Aedes aegypti and Aedes albopictus mosquitoes. CHIKV illness is characterized by fever and long-lasting arthritic symptoms, and in some cases it is a deadly disease. The CHIKV envelope E2 (E2CHIKV) glycoprotein is crucial for virus attachment to the cell. Furthermore, E2CHIKV is the immunodominant protein and the main target of neutralizing antibodies. To date, there is no available prophylactic vaccine or specific treatment against CHIKV infection. Here, we designed and produced a DNA vaccine and a recombinant protein containing a consensus sequence of E2CHIKV. C57BL/6 mice immunized twice with the E2CHIKV recombinant protein in the presence of the adjuvant Poly (I:C) induced the highest E2CHIKV-specific humoral and cellular immune responses, while the immunization with the homologous DNA vaccine pVAX-E2CHIKV was able to induce specific IFN-γ producing cells. The heterologous prime-boost strategy was also able to induce specific cellular and humoral immune responses that were, in general, lower than the responses induced by the homologous E2CHIKV recombinant protein immunization. Furthermore, recombinant E2CHIKV induced the highest titers of neutralizing antibodies. Collectively, we believe this is the first report to analyze E2CHIKV-specific humoral and cellular immune responses after immunization with E2CHIKV recombinant protein and DNA pVAX-E2CHIKV vaccine platforms.
{"title":"Prime-boost with Chikungunya virus E2 envelope protein combined with Poly (I:C) induces specific humoral and cellular immune responses","authors":"Marcelo Pires Amaral , Fernanda Caroline Coirada , Juliana de Souza Apostolico , Nádia Tomita , Edgar Ruz Fernandes , Higo Fernando Santos Souza , Rosa Maria Chura-Chambi , Ligia Morganti , Silvia Beatriz Boscardin , Daniela Santoro Rosa","doi":"10.1016/j.crimmu.2021.03.001","DOIUrl":"10.1016/j.crimmu.2021.03.001","url":null,"abstract":"<div><p>Chikungunya virus (CHIKV) is an arbovirus transmitted to humans mainly by the bite of infected <em>Aedes aegypti</em> and <em>Aedes albopictus</em> mosquitoes. CHIKV illness is characterized by fever and long-lasting arthritic symptoms, and in some cases it is a deadly disease. The CHIKV envelope E2 (E2<sub>CHIKV</sub>) glycoprotein is crucial for virus attachment to the cell. Furthermore, E2<sub>CHIKV</sub> is the immunodominant protein and the main target of neutralizing antibodies. To date, there is no available prophylactic vaccine or specific treatment against CHIKV infection. Here, we designed and produced a DNA vaccine and a recombinant protein containing a consensus sequence of E2<sub>CHIKV</sub>. C57BL/6 mice immunized twice with the E2<sub>CHIKV</sub> recombinant protein in the presence of the adjuvant Poly (I:C) induced the highest E2<sub>CHIKV</sub>-specific humoral and cellular immune responses, while the immunization with the homologous DNA vaccine pVAX-E2<sub>CHIKV</sub> was able to induce specific IFN-γ producing cells. The heterologous prime-boost strategy was also able to induce specific cellular and humoral immune responses that were, in general, lower than the responses induced by the homologous E2<sub>CHIKV</sub> recombinant protein immunization. Furthermore, recombinant E2<sub>CHIKV</sub> induced the highest titers of neutralizing antibodies. Collectively, we believe this is the first report to analyze E2<sub>CHIKV</sub>-specific humoral and cellular immune responses after immunization with E2<sub>CHIKV</sub> recombinant protein and DNA pVAX-E2<sub>CHIKV</sub> vaccine platforms.</p></div>","PeriodicalId":72750,"journal":{"name":"Current research in immunology","volume":"2 ","pages":"Pages 23-31"},"PeriodicalIF":0.0,"publicationDate":"2021-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.crimmu.2021.03.001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"48458763","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
DNA methylation plays a crucial role in polarising naïve lymphocytes towards their various sub-populations to fight against many immune challenges including establishment of tumour. Gamma-tocotrienol (γT3) is a natural form of vitamin E, reported to possess anticancer and immunomodulatory effects. This study reports the anticancer effects of γT3 through modulation of DNA methylation in several genes in CD4+ T-lymphocytes using a syngeneic mouse model of breast cancer. Female BALB/c mice were fed with γT3 or vehicle (soy oil) for two-weeks via oral gavage before they were inoculated with 4T1 mouse mammary cancer cells. Supplementation continued until the mice were sacrificed. At autopsy, blood was collected via cardiac puncture and CD4+ T-cells were isolated for DNA extraction. The DNA was analysed using the EpiTech Methyl II mouse T-helper cell differentiation PCR array. γT3 supplementation reduced tumour growth in the tumour-induced animals and modulated host immune system by inducing changes in DNA methylation patterns of the HOXA10, IRF4 and RORα genes, which are involved in differentiation and clonal expansion of CD4+ T-cells. Results suggest that γT3 may enhance cell-mediated immune response in mice with breast cancer by inducing changes in DNA methylation pattern.
{"title":"Gamma-tocotrienol modifies methylation of HOXA10, IRF4 and RORα genes in CD4+ T-lymphocytes: Evidence from a syngeneic mouse model of breast cancer","authors":"Ammu K. Radhakrishnan , Jeya Seela Anandha Rao , Shonia Subramaniam , Premdass Ramdas","doi":"10.1016/j.crimmu.2021.10.001","DOIUrl":"10.1016/j.crimmu.2021.10.001","url":null,"abstract":"<div><p>DNA methylation plays a crucial role in polarising naïve lymphocytes towards their various sub-populations to fight against many immune challenges including establishment of tumour. Gamma-tocotrienol (γT3) is a natural form of vitamin E, reported to possess anticancer and immunomodulatory effects. This study reports the anticancer effects of γT3 through modulation of DNA methylation in several genes in CD4<sup>+</sup> T-lymphocytes using a syngeneic mouse model of breast cancer. Female BALB/c mice were fed with γT3 or vehicle (soy oil) for two-weeks via oral gavage before they were inoculated with 4T1 mouse mammary cancer cells. Supplementation continued until the mice were sacrificed. At autopsy, blood was collected via cardiac puncture and CD4<sup>+</sup> T-cells were isolated for DNA extraction. The DNA was analysed using the EpiTech Methyl II mouse T-helper cell differentiation PCR array. γT3 supplementation reduced tumour growth in the tumour-induced animals and modulated host immune system by inducing changes in DNA methylation patterns of the <em>HOXA10</em>, <em>IRF4</em> and <em>RORα</em> genes, which are involved in differentiation and clonal expansion of CD4<sup>+</sup> T-cells. Results suggest that γT3 may enhance cell-mediated immune response in mice with breast cancer by inducing changes in DNA methylation pattern.</p></div>","PeriodicalId":72750,"journal":{"name":"Current research in immunology","volume":"2 ","pages":"Pages 169-174"},"PeriodicalIF":0.0,"publicationDate":"2021-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2590255521000184/pdfft?md5=28398a743fbd4174366cc98d31ce4c2c&pid=1-s2.0-S2590255521000184-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41662044","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-01-01DOI: 10.1016/j.crimmu.2021.02.001
Jadesola (Jadé) Temitope Olayinka , Jillian M. Richmond
Alopecia areata (AA) is an autoimmune disorder resulting in hair loss. It has numerous variants or patterns, including diffuse type, patchy type, AA totalis, AA universalis, and more. In this graphical review, we provide an overview of AA immunopathogenesis, highlighting loss of immune privilege in the hair follicle as well as key immune cell types, cytokines and chemokines that drive autoimmune attack of the hair follicle. We also summarize recent literature identifying agents that block these pathways that could serve as new immunomodulatory treatments for AA.
{"title":"Immunopathogenesis of alopecia areata","authors":"Jadesola (Jadé) Temitope Olayinka , Jillian M. Richmond","doi":"10.1016/j.crimmu.2021.02.001","DOIUrl":"10.1016/j.crimmu.2021.02.001","url":null,"abstract":"<div><p>Alopecia areata (AA) is an autoimmune disorder resulting in hair loss. It has numerous variants or patterns, including diffuse type, patchy type, AA totalis, AA universalis, and more. In this graphical review, we provide an overview of AA immunopathogenesis, highlighting loss of immune privilege in the hair follicle as well as key immune cell types, cytokines and chemokines that drive autoimmune attack of the hair follicle. We also summarize recent literature identifying agents that block these pathways that could serve as new immunomodulatory treatments for AA.</p></div>","PeriodicalId":72750,"journal":{"name":"Current research in immunology","volume":"2 ","pages":"Pages 7-11"},"PeriodicalIF":0.0,"publicationDate":"2021-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.crimmu.2021.02.001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"44649812","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-01-01DOI: 10.1016/j.crimmu.2021.10.004
Patrick Schriek , Haiyin Liu , Alan C. Ching , Pauline Huang , Nishma Gupta , Kayla R. Wilson , MinHsuang Tsai , Yuting Yan , Christophe F. Macri , Laura F. Dagley , Giuseppe Infusini , Andrew I. Webb , Hamish E.G. McWilliam , Satoshi Ishido , Justine D. Mintern , Jose A. Villadangos
MARCH1 and MARCH8 are ubiquitin ligases that control the expression and trafficking of critical immunoreceptors. Understanding of their function is hampered by three major knowledge gaps: (i) it is unclear which cell types utilize these ligases; (ii) their level of redundancy is unknown; and (iii) most of their putative substrates have been described in cell lines, often overexpressing MARCH1 or MARCH8, and it is unclear which substrates are regulated by either ligase in vivo. Here we address these questions by systematically analyzing the immune cell repertoire of MARCH1- or MARCH8-deficient mice, and applying unbiased proteomic profiling of the plasma membrane of primary cells to identify MARCH1 and MARCH8 substrates. Only CD86 and MHC II were unequivocally identified as immunoreceptors regulated by MARCH1 and MARCH8, but each ligase carried out its function in different tissues. MARCH1 regulated MHC II and CD86 in professional and “atypical” antigen presenting cells of hematopoietic origin, including neutrophils, eosinophils and monocytes. MARCH8 only operated in non-hematopoietic cells, such as thymic and alveolar epithelial cells. Our results establish the tissue-specific functions of MARCH1 and MARCH8 in regulation of immune receptor expression and reveal that the range of cells constitutively endowed with antigen-presentation capacity is wider than generally appreciated.
{"title":"Physiological substrates and ontogeny-specific expression of the ubiquitin ligases MARCH1 and MARCH8","authors":"Patrick Schriek , Haiyin Liu , Alan C. Ching , Pauline Huang , Nishma Gupta , Kayla R. Wilson , MinHsuang Tsai , Yuting Yan , Christophe F. Macri , Laura F. Dagley , Giuseppe Infusini , Andrew I. Webb , Hamish E.G. McWilliam , Satoshi Ishido , Justine D. Mintern , Jose A. Villadangos","doi":"10.1016/j.crimmu.2021.10.004","DOIUrl":"10.1016/j.crimmu.2021.10.004","url":null,"abstract":"<div><p>MARCH1 and MARCH8 are ubiquitin ligases that control the expression and trafficking of critical immunoreceptors. Understanding of their function is hampered by three major knowledge gaps: (i) it is unclear which cell types utilize these ligases; (ii) their level of redundancy is unknown; and (iii) most of their putative substrates have been described in cell lines, often overexpressing MARCH1 or MARCH8, and it is unclear which substrates are regulated by either ligase <em>in vivo</em>. Here we address these questions by systematically analyzing the immune cell repertoire of MARCH1- or MARCH8-deficient mice, and applying unbiased proteomic profiling of the plasma membrane of primary cells to identify MARCH1 and MARCH8 substrates. Only CD86 and MHC II were unequivocally identified as immunoreceptors regulated by MARCH1 and MARCH8, but each ligase carried out its function in different tissues. MARCH1 regulated MHC II and CD86 in professional and “atypical” antigen presenting cells of hematopoietic origin, including neutrophils, eosinophils and monocytes. MARCH8 only operated in non-hematopoietic cells, such as thymic and alveolar epithelial cells. Our results establish the tissue-specific functions of MARCH1 and MARCH8 in regulation of immune receptor expression and reveal that the range of cells constitutively endowed with antigen-presentation capacity is wider than generally appreciated.</p></div>","PeriodicalId":72750,"journal":{"name":"Current research in immunology","volume":"2 ","pages":"Pages 218-228"},"PeriodicalIF":0.0,"publicationDate":"2021-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2590255521000214/pdfft?md5=9a6d9a84806acc387c9644dde358269c&pid=1-s2.0-S2590255521000214-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"42664022","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-01-01DOI: 10.1016/j.crimmu.2021.12.001
Simone Morris , Kathryn Wright , Vamshikrishna Malyla , Warwick J. Britton , Philip M. Hansbro , Pradeep Manuneedhi Cholan , Stefan H. Oehlers
Cigarette smoke (CS)-induced inflammation leads to a range of diseases including chronic obstructive pulmonary disease and cancer. The gut microbiota is a major modifying environmental factor that determine the severity of cigarette smoke-induced pathology. Microbiomes and metabolites from CS-exposed mice exacerbate lung inflammation via the gut-lung axis of shared mucosal immunity in mice but these systems are expensive to establish and analyse. Zebrafish embryos and larvae have been used to model the effects of cigarette smoking on a range of physiological processes and offer an amenable platform for screening modifiers of cigarette smoke-induced pathologies with key features of low cost and rapid visual readouts. Here we exposed zebrafish larvae to cigarette smoke extract (CSE) and characterised a CSE-induced leukocytic inflammatory phenotype with increased neutrophilic and macrophage inflammation in the gut. The CSE-induced phenotype was exacerbated by co-exposure to microbiota from the faeces of CS-exposed mice, but not control mice. Microbiota could be recovered from the gut of zebrafish and studied in isolation in a screening setting. This demonstrates the utility of the zebrafish-CSE exposure platform for identifying environmental modifiers of cigarette smoking-associated pathology and demonstrates that the CS-exposed mouse gut microbiota potentiates the inflammatory effects of CSE across host species.
{"title":"Exposure to the gut microbiota from cigarette smoke-exposed mice exacerbates cigarette smoke extract-induced inflammation in zebrafish larvae","authors":"Simone Morris , Kathryn Wright , Vamshikrishna Malyla , Warwick J. Britton , Philip M. Hansbro , Pradeep Manuneedhi Cholan , Stefan H. Oehlers","doi":"10.1016/j.crimmu.2021.12.001","DOIUrl":"10.1016/j.crimmu.2021.12.001","url":null,"abstract":"<div><p>Cigarette smoke (CS)-induced inflammation leads to a range of diseases including chronic obstructive pulmonary disease and cancer. The gut microbiota is a major modifying environmental factor that determine the severity of cigarette smoke-induced pathology. Microbiomes and metabolites from CS-exposed mice exacerbate lung inflammation via the gut-lung axis of shared mucosal immunity in mice but these systems are expensive to establish and analyse. Zebrafish embryos and larvae have been used to model the effects of cigarette smoking on a range of physiological processes and offer an amenable platform for screening modifiers of cigarette smoke-induced pathologies with key features of low cost and rapid visual readouts. Here we exposed zebrafish larvae to cigarette smoke extract (CSE) and characterised a CSE-induced leukocytic inflammatory phenotype with increased neutrophilic and macrophage inflammation in the gut. The CSE-induced phenotype was exacerbated by co-exposure to microbiota from the faeces of CS-exposed mice, but not control mice. Microbiota could be recovered from the gut of zebrafish and studied in isolation in a screening setting. This demonstrates the utility of the zebrafish-CSE exposure platform for identifying environmental modifiers of cigarette smoking-associated pathology and demonstrates that the CS-exposed mouse gut microbiota potentiates the inflammatory effects of CSE across host species.</p></div>","PeriodicalId":72750,"journal":{"name":"Current research in immunology","volume":"2 ","pages":"Pages 229-236"},"PeriodicalIF":0.0,"publicationDate":"2021-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S259025552100024X/pdfft?md5=e6b2c1f1945d4e37d9734ad0c142d08c&pid=1-s2.0-S259025552100024X-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"45004075","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-01-01DOI: 10.1016/j.crimmu.2021.07.002
Katrien Deroost , Christopher Alder , Caroline Hosking, Sarah McLaughlin, Jing-Wen Lin , Matthew D. Lewis, Yolanda Saavedra-Torres, John W.G. Addy, Prisca Levy, Maria Giorgalli, Jean Langhorne
Natural infection with Plasmodium parasites, the causative agents of malaria, occurs via mosquito vectors. However, most of our knowledge of the immune response to the blood stages of Plasmodium is from infections initiated by injection of serially blood-passaged infected red blood cells, resulting in an incomplete life cycle in the mammalian host. Vector transmission of the rodent malaria parasite, Plasmodium chabaudi chabaudi AS has been shown to give rise to a more attenuated blood-stage infection in C57Bl/6J mice, when compared to infections initiated with serially blood-passaged P. chabaudi-infected red blood cells. In mouse models, the host immune response induced by parasites derived from natural mosquito transmission is likely to more closely resemble the immune responses to Plasmodium infections in humans. It is therefore important to determine how the host response differs between the two types of infections.
As the spleen is considered to be a major contributor to the protective host response to P. chabaudi, we carried out a comparative transcriptomic analysis of the splenic response to recently mosquito-transmitted and serially blood-passaged parasites in C57Bl/6J mice. The attenuated infection arising from recently mosquito-transmitted parasites is characterised by an earlier and stronger myeloid- and IFNγ-related response. Analyses of spleen lysates from the two infections similarly showed stronger or earlier inflammatory cytokine and chemokine production in the recently mosquito-transmitted blood-stage infections. Furthermore, tissue macrophages, including red pulp macrophages, and IFNγ-signalling in myeloid cells, are required for the early control of P. chabaudi recently mosquito-transmitted parasites, thus contributing to the attenuation of mosquito-transmitted infections.
The molecules responsible for this early activation response to recently-transmitted blood-stage parasites in mice would be important to identify, as they may help to elucidate the nature of the initial interactions between blood-stage parasites and the host immune system in naturally transmitted malaria.
{"title":"Tissue macrophages and interferon-gamma signalling control blood-stage Plasmodium chabaudi infections derived from mosquito-transmitted parasites","authors":"Katrien Deroost , Christopher Alder , Caroline Hosking, Sarah McLaughlin, Jing-Wen Lin , Matthew D. Lewis, Yolanda Saavedra-Torres, John W.G. Addy, Prisca Levy, Maria Giorgalli, Jean Langhorne","doi":"10.1016/j.crimmu.2021.07.002","DOIUrl":"10.1016/j.crimmu.2021.07.002","url":null,"abstract":"<div><p>Natural infection with <em>Plasmodium</em> parasites, the causative agents of malaria, occurs via mosquito vectors. However, most of our knowledge of the immune response to the blood stages of <em>Plasmodium</em> is from infections initiated by injection of serially blood-passaged infected red blood cells, resulting in an incomplete life cycle in the mammalian host. Vector transmission of the rodent malaria parasite, <em>Plasmodium chabaudi chabaudi</em> AS has been shown to give rise to a more attenuated blood-stage infection in C57Bl/6J mice, when compared to infections initiated with serially blood-passaged <em>P. chabaudi</em>-infected red blood cells. In mouse models, the host immune response induced by parasites derived from natural mosquito transmission is likely to more closely resemble the immune responses to <em>Plasmodium</em> infections in humans. It is therefore important to determine how the host response differs between the two types of infections.</p><p>As the spleen is considered to be a major contributor to the protective host response to <em>P. chabaudi</em>, we carried out a comparative transcriptomic analysis of the splenic response to recently mosquito-transmitted and serially blood-passaged parasites in C57Bl/6J mice. The attenuated infection arising from recently mosquito-transmitted parasites is characterised by an earlier and stronger myeloid- and IFNγ-related response. Analyses of spleen lysates from the two infections similarly showed stronger or earlier inflammatory cytokine and chemokine production in the recently mosquito-transmitted blood-stage infections. Furthermore, tissue macrophages, including red pulp macrophages, and IFNγ-signalling in myeloid cells, are required for the early control of <em>P. chabaudi</em> recently mosquito-transmitted parasites, thus contributing to the attenuation of mosquito-transmitted infections.</p><p>The molecules responsible for this early activation response to recently-transmitted blood-stage parasites in mice would be important to identify, as they may help to elucidate the nature of the initial interactions between blood-stage parasites and the host immune system in naturally transmitted malaria.</p></div>","PeriodicalId":72750,"journal":{"name":"Current research in immunology","volume":"2 ","pages":"Pages 104-119"},"PeriodicalIF":0.0,"publicationDate":"2021-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8428512/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39425923","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-01-01DOI: 10.1016/j.crimmu.2021.09.001
Carlo Lombardi , Elena Roca , Barbara Bigni , Bruno Bertozzi , Camillo Ferrandina , Alberto Franzin , Oscar Vivaldi , Marcello Cottini , Andrea D'Alessio , Paolo Del Poggio , Gian Marco Conte , Alvise Berti
Early prediction of COVID-19 in-hospital mortality relies usually on patients’ preexisting comorbidities and is rarely reproducible in independent cohorts. We wanted to compare the role of routinely measured biomarkers of immunity, inflammation, and cellular damage with preexisting comorbidities in eight different machine-learning models to predict mortality, and evaluate their performance in an independent population. We recruited and followed-up consecutive adult patients with SARS-Cov-2 infection in two different Italian hospitals. We predicted 60-day mortality in one cohort (development dataset, n = 299 patients, of which 80% was allocated to the development dataset and 20% to the training set) and retested the models in the second cohort (external validation dataset, n = 402).
Demographic, clinical, and laboratory features at admission, treatments and disease outcomes were significantly different between the two cohorts. Notably, significant differences were observed for %lymphocytes (p < 0.05), international-normalized-ratio (p < 0.01), platelets, alanine-aminotransferase, creatinine (all p < 0.001). The primary outcome (60-day mortality) was 29.10% (n = 87) in the development dataset, and 39.55% (n = 159) in the external validation dataset. The performance of the 8 tested models on the external validation dataset were similar to that of the holdout test dataset, indicating that the models capture the key predictors of mortality. The shap analysis in both datasets showed that age, immune features (%lymphocytes, platelets) and LDH substantially impacted on all models' predictions, while creatinine and CRP varied among the different models. The model with the better performance was model 8 (60-day mortality AUROC 0.83 ± 0.06 in holdout test set, 0.79 ± 0.02 in external validation dataset). The features that had the greatest impact on this model's prediction were age, LDH, platelets, and %lymphocytes, more than comorbidities or inflammation markers, and these findings were highly consistent in both datasets, likely reflecting the virus effect at the very beginning of the disease.
{"title":"Immune and cellular damage biomarkers to predict COVID-19 mortality in hospitalized patients","authors":"Carlo Lombardi , Elena Roca , Barbara Bigni , Bruno Bertozzi , Camillo Ferrandina , Alberto Franzin , Oscar Vivaldi , Marcello Cottini , Andrea D'Alessio , Paolo Del Poggio , Gian Marco Conte , Alvise Berti","doi":"10.1016/j.crimmu.2021.09.001","DOIUrl":"10.1016/j.crimmu.2021.09.001","url":null,"abstract":"<div><p>Early prediction of COVID-19 in-hospital mortality relies usually on patients’ preexisting comorbidities and is rarely reproducible in independent cohorts. We wanted to compare the role of routinely measured biomarkers of immunity, inflammation, and cellular damage with preexisting comorbidities in eight different machine-learning models to predict mortality, and evaluate their performance in an independent population. We recruited and followed-up consecutive adult patients with SARS-Cov-2 infection in two different Italian hospitals. We predicted 60-day mortality in one cohort (development dataset, n = 299 patients, of which 80% was allocated to the development dataset and 20% to the training set) and retested the models in the second cohort (external validation dataset, n = 402).</p><p>Demographic, clinical, and laboratory features at admission, treatments and disease outcomes were significantly different between the two cohorts. Notably, significant differences were observed for %lymphocytes (p < 0.05), international-normalized-ratio (p < 0.01), platelets, alanine-aminotransferase, creatinine (all p < 0.001). The primary outcome (60-day mortality) was 29.10% (n = 87) in the development dataset, and 39.55% (n = 159) in the external validation dataset. The performance of the 8 tested models on the external validation dataset were similar to that of the holdout test dataset, indicating that the models capture the key predictors of mortality. The shap analysis in both datasets showed that age, immune features (%lymphocytes, platelets) and LDH substantially impacted on all models' predictions, while creatinine and CRP varied among the different models. The model with the better performance was model 8 (60-day mortality AUROC 0.83 ± 0.06 in holdout test set, 0.79 ± 0.02 in external validation dataset). The features that had the greatest impact on this model's prediction were age, LDH, platelets, and %lymphocytes, more than comorbidities or inflammation markers, and these findings were highly consistent in both datasets, likely reflecting the virus effect at the very beginning of the disease.</p></div>","PeriodicalId":72750,"journal":{"name":"Current research in immunology","volume":"2 ","pages":"Pages 155-162"},"PeriodicalIF":0.0,"publicationDate":"2021-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/2e/d6/main.PMC8444380.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39434719","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-01-01DOI: 10.1016/j.crimmu.2021.06.001
Amrita Mishra, Girdhari Lal
Neurokinin receptors belong to the GPCRs family and are ubiquitously expressed throughout the nervous and immune systems. Neurokinin receptors in coordination with neurokinins playing an important role in many physiological processes, including smooth muscle contraction, secretion, proliferation, and nociception. They also contribute to various disease conditions such as inflammatory bowel disease, rheumatoid arthritis, multiple sclerosis, psoriasis, and cancer. Neurokinin receptors antagonist are potent and highly selective and showing success in treating chemotherapy-induced nausea and vomiting. In this review, discuss the various neurokinin receptor expression on immune cells and their importance in various inflammatory and autoimmune diseases and their therapeutic importance.
{"title":"Neurokinin receptors and their implications in various autoimmune diseases","authors":"Amrita Mishra, Girdhari Lal","doi":"10.1016/j.crimmu.2021.06.001","DOIUrl":"10.1016/j.crimmu.2021.06.001","url":null,"abstract":"<div><p>Neurokinin receptors belong to the GPCRs family and are ubiquitously expressed throughout the nervous and immune systems. Neurokinin receptors in coordination with neurokinins playing an important role in many physiological processes, including smooth muscle contraction, secretion, proliferation, and nociception. They also contribute to various disease conditions such as inflammatory bowel disease, rheumatoid arthritis, multiple sclerosis, psoriasis, and cancer. Neurokinin receptors antagonist are potent and highly selective and showing success in treating chemotherapy-induced nausea and vomiting. In this review, discuss the various neurokinin receptor expression on immune cells and their importance in various inflammatory and autoimmune diseases and their therapeutic importance.</p></div>","PeriodicalId":72750,"journal":{"name":"Current research in immunology","volume":"2 ","pages":"Pages 66-78"},"PeriodicalIF":0.0,"publicationDate":"2021-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.crimmu.2021.06.001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49190248","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2021-01-01DOI: 10.1016/j.crimmu.2021.10.002
Wilson Mandala , Visopo Harawa , Alinane Munyenyembe , Monica Soko , Herbert Longwe
Cell-mediated responses to immunological stimuli are often localised in inflammatory sites and involve a number of cell types. These responses can be functionally characterised at the single-cell level on the basis of the types of cytokines expressed either in whole blood or PBMCs. The ability to measure antigen-specific cell responses at the single cell level is an important tool with a wide range of potential applications ranging from studies of disease pathogenesis to the evaluation of vaccines.
A number of experiments were performed in this study in order to establish the optimal conditions for in vitro stimulation of cytokine production by T cells and monocytes in whole blood samples collected from healthy adult Malawian participants and the optimal staining conditions for various cytokine producing cells. Different stimulation methods and conditions, different culture tubes and incubators and different antibody labelling conditions were assessed in order to establish optimal conditions for detecting cytokine-producing cells in whole blood samples.
The use of PMA plus Ionomycin produced highest cytokine-producing T cells whereas LPS was a better stimulant for cytokine producing monocytes. Stimulation of whole blood for 5 h was optimal for cytokine detection in T cells whereas 4 h was optimal for monocytes. BFA was found to be a better Golgi blocker than Monensin and the use of 15 ml Falcon-type polypropylene tubes while stationary resulted in the detection of the highest proportion of cytokine-producing cells. T cells were found to be producers of mainly TNF-α, IFN-γ and IL-2 whereas Monocytes were mainly producing TNF-α and IL-6. Anti-CD3-PerCP (used at a ratio of 1:25), anti-CD14-APC (used at a ratio of 1:50) and anti-cytokine-PE (used at a ratio of 1:12.5) resulted in the best results. The highest cytokine production monocytes were detected when 1 X FACS Lysing solution was used at a volume of 40X that of the whole blood sample compared to the other volumes. These optimal conditions are essential in determination of proportion of cytokine-producing cells using ICS in whole blood.
{"title":"Optimization of stimulation and staining conditions for intracellular cytokine staining (ICS) for determination of cytokine-producing T cells and monocytes","authors":"Wilson Mandala , Visopo Harawa , Alinane Munyenyembe , Monica Soko , Herbert Longwe","doi":"10.1016/j.crimmu.2021.10.002","DOIUrl":"10.1016/j.crimmu.2021.10.002","url":null,"abstract":"<div><p>Cell-mediated responses to immunological stimuli are often localised in inflammatory sites and involve a number of cell types. These responses can be functionally characterised at the single-cell level on the basis of the types of cytokines expressed either in whole blood or PBMCs. The ability to measure antigen-specific cell responses at the single cell level is an important tool with a wide range of potential applications ranging from studies of disease pathogenesis to the evaluation of vaccines.</p><p>A number of experiments were performed in this study in order to establish the optimal conditions for <em>in vitro</em> stimulation of cytokine production by T cells and monocytes in whole blood samples collected from healthy adult Malawian participants and the optimal staining conditions for various cytokine producing cells. Different stimulation methods and conditions, different culture tubes and incubators and different antibody labelling conditions were assessed in order to establish optimal conditions for detecting cytokine-producing cells in whole blood samples.</p><p>The use of PMA plus Ionomycin produced highest cytokine-producing T cells whereas LPS was a better stimulant for cytokine producing monocytes. Stimulation of whole blood for 5 h was optimal for cytokine detection in T cells whereas 4 h was optimal for monocytes. BFA was found to be a better Golgi blocker than Monensin and the use of 15 ml Falcon-type polypropylene tubes while stationary resulted in the detection of the highest proportion of cytokine-producing cells. T cells were found to be producers of mainly TNF-α, IFN-γ and IL-2 whereas Monocytes were mainly producing TNF-α and IL-6. Anti-CD3-PerCP (used at a ratio of 1:25), anti-CD14-APC (used at a ratio of 1:50) and anti-cytokine-PE (used at a ratio of 1:12.5) resulted in the best results. The highest cytokine production monocytes were detected when 1 X FACS Lysing solution was used at a volume of 40X that of the whole blood sample compared to the other volumes. These optimal conditions are essential in determination of proportion of cytokine-producing cells using ICS in whole blood.</p></div>","PeriodicalId":72750,"journal":{"name":"Current research in immunology","volume":"2 ","pages":"Pages 184-193"},"PeriodicalIF":0.0,"publicationDate":"2021-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2590255521000196/pdfft?md5=1c5ea2e935532d6ad83f82c3e1b11aa0&pid=1-s2.0-S2590255521000196-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"48874425","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}