Pub Date : 2023-11-22DOI: 10.1016/j.cellimm.2023.104788
Jeffrey Maslanka , Gretel Torres , Jennifer Londregan , Naomi Goldman , Daniel Silberman , John Somerville, James E. Riggs
Recent advances in immunotherapy have not addressed the challenge presented by ovarian cancer. Although the peritoneum is an “accessible” locus for this disease there has been limited characterization of the immunobiology therein. We investigated the ID8-C57BL/6J ovarian cancer model and found marked depletion of B1 cells from the ascites of the peritoneal cavity. There was also selective loss of the B1 and marginal zone B cell subsets from the spleen. Immunity to antigens that activate these subsets validated their loss rather than relocation. A marked influx of myeloid-derived suppressor cells correlated with B cell subset depletion. These observations are discussed in the context of the housekeeping burden placed on innate B cells during ovarian cancer and to foster consideration of B cell biology in therapeutic strategies to address this challenge.
{"title":"Loss of B1 and marginal zone B cells during ovarian cancer","authors":"Jeffrey Maslanka , Gretel Torres , Jennifer Londregan , Naomi Goldman , Daniel Silberman , John Somerville, James E. Riggs","doi":"10.1016/j.cellimm.2023.104788","DOIUrl":"https://doi.org/10.1016/j.cellimm.2023.104788","url":null,"abstract":"<div><p><span>Recent advances in immunotherapy<span> have not addressed the challenge presented by ovarian cancer. Although the peritoneum is an “accessible” locus for this disease there has been limited characterization of the immunobiology<span> therein. We investigated the ID8-C57BL/6J ovarian cancer model and found marked depletion of B1 cells from the ascites of the peritoneal cavity. There was also selective loss of the B1 and </span></span></span>marginal zone B cell<span> subsets from the spleen. Immunity to antigens that activate these subsets validated their loss rather than relocation. A marked influx of myeloid-derived suppressor cells correlated with B cell subset depletion. These observations are discussed in the context of the housekeeping burden placed on innate B cells during ovarian cancer and to foster consideration of B cell biology in therapeutic strategies to address this challenge.</span></p></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"395 ","pages":"Article 104788"},"PeriodicalIF":4.3,"publicationDate":"2023-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138423658","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-11-01DOI: 10.1016/j.cellimm.2023.104784
Lili Liu , Yateng Li , Yu Song , Zhen Sun , Wenjing Li , Bin Li , Yongjie Wang , Haibo Wang , Bin Wang
Antigenic peptides play a central role in immune surveillance in cancer, infectious disease, autoimmunity, and allergy. The identification and isolation of antigenic peptides for T cell immune response are crucial for successful personalized adoptive immune cell therapy. The mainly methods includes gene sequencing and bioinformatic analysis. The antigenic peptides which identified by analysis and artificially synthesized still need antigen presenting cell (APC) to deliver to T cells. However, high costs and lengthy process times have limited its application in clinical practice. In order to overcome it, this study attempted to directly capture antigenic peptide-major histocompatibility complex (MHC) class I (pMHCs) from cell lysates using streptavidin Dynabeads and biotin-labeled antibodies, then the pMHCs was co-cultured with tumor infiltrating lymphocytes (TILs) of the same tissue origin. The results indicated that the captured pMHCs were able to enrich the tumor antigen-specific CD8+ T cells, and also effectively induce proliferation and cytotoxic responses of CD8+ T cells. This study provided a novel approach for obtaining tumor antigenic pMHCs, which could enrich antigen-specific CD8+ T cells, and could also function as artificial APCs (aAPCs) to stimulate proliferation and activation of T cells. Notably, these pMHCs can stimulate the proliferation of stem-like memory T cells. In conclusion, this study describes a time-saving and low-cost method to isolate tumour antigen peptide MHC complexs, helping tumor antigen-specific T cell enrichment, activation, and proliferation.
{"title":"One-step shotgun approach for antigenic specific pMHCs capture stimulated CD8+ T cell activation and proliferation","authors":"Lili Liu , Yateng Li , Yu Song , Zhen Sun , Wenjing Li , Bin Li , Yongjie Wang , Haibo Wang , Bin Wang","doi":"10.1016/j.cellimm.2023.104784","DOIUrl":"10.1016/j.cellimm.2023.104784","url":null,"abstract":"<div><p>Antigenic peptides play a central role in immune surveillance in cancer, infectious disease, autoimmunity, and allergy. The identification and isolation of antigenic peptides for T cell immune response are crucial for successful personalized adoptive immune cell therapy. The mainly methods includes gene sequencing and bioinformatic analysis. The antigenic peptides which identified by analysis and artificially synthesized still need antigen presenting cell (APC) to deliver to T cells. However, high costs and lengthy process times have limited its application in clinical practice. In order to overcome it, this study attempted to directly capture antigenic peptide-major histocompatibility complex (MHC) class I (pMHCs) from cell lysates using streptavidin Dynabeads and biotin-labeled antibodies, then the pMHCs was co-cultured with tumor infiltrating lymphocytes (TILs) of the same tissue origin. The results indicated that the captured pMHCs were able to enrich the tumor antigen-specific CD8<sup>+</sup> T cells, and also effectively induce proliferation and cytotoxic responses of CD8<sup>+</sup> T cells. This study provided a novel approach for obtaining tumor antigenic pMHCs, which could enrich antigen-specific CD8<sup>+</sup> T cells, and could also function as artificial APCs (aAPCs) to stimulate proliferation and activation of T cells. Notably, these pMHCs can stimulate the proliferation of stem-like memory T cells. In conclusion, this study describes a time-saving and low-cost method to isolate tumour antigen peptide MHC complexs, helping tumor antigen-specific T cell enrichment, activation, and proliferation.</p></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"393 ","pages":"Article 104784"},"PeriodicalIF":4.3,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"135566336","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Regulatory T (Treg) cells interact with a variety of resident cells and infiltrated immune cells in the central nervous system (CNS) to modulate neuroinflammation and neurodegeneration. Extracellular amyloid-β (Aβ) peptide deposition and secondary persistent inflammation due to activation of microglia, astrocytes, and infiltrated immune cells contribute to Alzheimer's disease (AD)-related neurodegeneration. The majority of evidence supports the neuroprotective effects of Treg cells in AD. In the early stages of AD, appropriate Treg cell activity is required for the induction of microglia and astrocyte phagocytic activity in order to clear A deposits and prevent neuroinflammation. Such neuroprotective impacts were in part attributed to the ability of Treg cells to suppress deleterious and/or boost beneficial functions of microglia/astrocytes. In the later stages of AD, an effective Treg cell activity needs to prevent neurotoxicity and neurodegeneration. Treg cells can exert preventive effects on Th1-, and Th17 cell-related pathologic responses, whilst potentiating Th2-mediated protective activity. The impaired Treg cell-related immunomodulatory mechanisms have been described in AD patients and in related animal models which can contribute to the onset and progression of AD. This review aimed to provide a comprehensive figure regarding the role of Treg cells in AD while highlighting potential therapeutic approaches.
{"title":"Differential roles of regulatory T cells in Alzheimer's disease","authors":"Abdollah Jafarzadeh , Abdolkarim Sheikhi , Zahra Jafarzadeh , Maryam Nemati","doi":"10.1016/j.cellimm.2023.104778","DOIUrl":"10.1016/j.cellimm.2023.104778","url":null,"abstract":"<div><p>Regulatory T (Treg) cells interact with a variety of resident cells and infiltrated immune cells in the central nervous system (CNS) to modulate neuroinflammation and neurodegeneration. Extracellular amyloid-β (Aβ) peptide deposition and secondary persistent inflammation due to activation of microglia, astrocytes, and infiltrated immune cells contribute to Alzheimer's disease (AD)-related neurodegeneration. The majority of evidence supports the neuroprotective effects of Treg cells in AD. In the early stages of AD, appropriate Treg cell activity is required for the induction of microglia and astrocyte phagocytic activity in order to clear A deposits and prevent neuroinflammation. Such neuroprotective impacts were in part attributed to the ability of Treg cells to suppress deleterious and/or boost beneficial functions of microglia/astrocytes. In the later stages of AD, an effective Treg cell activity needs to prevent neurotoxicity and neurodegeneration. Treg cells can exert preventive effects on Th1-, and Th17 cell-related pathologic responses, whilst potentiating Th2-mediated protective activity. The impaired Treg cell-related immunomodulatory mechanisms have been described in AD patients and in related animal models which can contribute to the onset and progression of AD. This review aimed to provide a comprehensive figure regarding the role of Treg cells in AD while highlighting potential therapeutic approaches.</p></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"393 ","pages":"Article 104778"},"PeriodicalIF":4.3,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71421055","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-11-01DOI: 10.1016/j.cellimm.2023.104782
Rui Wang , Tao Zhang , Yuan Lu , Yalong Lin , Shuyuan Kou , Xuefeng Li , Yang Wang , Liangzhi Xie
Type I interferons (IFN), especially human IFN alpha (IFNα), have been utilized for antitumor therapy for decades. Human interferon beta (IFNβ) is rarely used for cancer treatment, despite advantages over IFNα in biological activities such as tumor growth inhibition and dendritic cell (DC) activation. The utilization of pegylated human IFNβ (PEG-IFNβ), as monotherapy or in combination with immune checkpoint inhibitors (ICIs) was evaluated in this study through in vivo efficacy studies in syngeneic mouse melanoma, non-small cell lung cancer (NSCLC), and colon adenocarcinoma (COAD) models resistant to immune checkpoint inhibitors (ICIs). In vitro comparative study of PEG-IFNβ and pegylated IFNα-2b was performed in terms of tumor growth inhibition against human melanoma, NSCLC and COAD cell lines and activation of human monocyte-derived DCs (MoDCs). Our data demonstrate that the in vivo antitumor effects of PEG-IFNβ are partially attributable to tumor growth-inhibitory effects and DC-activating activities, superior to pegylated IFNα-2b. Our findings suggest that utilizing PEG-IFNβ as an antitumor therapy can enhance the therapeutic effect of ICIs in ICI-resistant tumors by directly inhibiting tumor growth and induction of DC maturation.
{"title":"Antitumor activity of pegylated human interferon β as monotherapy or in combination with immune checkpoint inhibitors via tumor growth inhibition and dendritic cell activation","authors":"Rui Wang , Tao Zhang , Yuan Lu , Yalong Lin , Shuyuan Kou , Xuefeng Li , Yang Wang , Liangzhi Xie","doi":"10.1016/j.cellimm.2023.104782","DOIUrl":"10.1016/j.cellimm.2023.104782","url":null,"abstract":"<div><p>Type I interferons (IFN), especially human IFN alpha (IFNα), have been utilized for antitumor therapy for decades. Human interferon beta (IFNβ) is rarely used for cancer treatment, despite advantages over IFNα in biological activities such as tumor growth inhibition and dendritic cell (DC) activation. The utilization of pegylated human IFNβ (PEG-IFNβ), as monotherapy or in combination with immune checkpoint inhibitors (ICIs) was evaluated in this study through in vivo efficacy studies in syngeneic mouse melanoma, non-small cell lung cancer (NSCLC), and colon adenocarcinoma (COAD) models resistant to immune checkpoint inhibitors (ICIs). In vitro comparative study of PEG-IFNβ and pegylated IFNα-2b was performed in terms of tumor growth inhibition against human melanoma, NSCLC and COAD cell lines and activation of human monocyte-derived DCs (MoDCs). Our data demonstrate that the in vivo antitumor effects of PEG-IFNβ are partially attributable to tumor growth-inhibitory effects and DC-activating activities, superior to pegylated IFNα-2b. Our findings suggest that utilizing PEG-IFNβ as an antitumor therapy can enhance the therapeutic effect of ICIs in ICI-resistant tumors by directly inhibiting tumor growth and induction of DC maturation.</p></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"393 ","pages":"Article 104782"},"PeriodicalIF":4.3,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71478442","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Inflammatory bowel diseases are associated with dysregulated inflammatory immune responses in the gastrointestinal tract. We found that deficiencies of both IL-4 receptor alpha chain (IL-4Rα) and IL-10 in BALB/c mice (IL-4Rα × IL-10 KO mice) highly induced spontaneous rectal prolapse and diarrhea. These mice also exhibited severe colitis in their cecum and colon and marked elevation of serum proinflammatory cytokines including TNFα and IFNγ. These pathologies were transmittable with their cecal contents containing Helicobacter spp. Their mesenteric LN cells produced TNFα and IFNγ in response to soluble H. hepaticus antigens and high titers of H. hepaticus-specific serum IgG were also detected. These results suggested the important function of IL-4Rα signaling in controlling the intestinal inflammation and the susceptibility to intestinal microbes including H. hepaticus. Therefore, these IL-4Rα × IL-10 KO mice potentially provide the significant murine model for clarifying the causes and control of spontaneous colitis and intestinal inflammation.
{"title":"Genetic deficiencies of both IL-4 receptor alpha chain and IL-10 trigger early onset of severe colitis in mice","authors":"Hisashi Nagase , Masaya Takamoto , Nancy Noben-Trauth","doi":"10.1016/j.cellimm.2023.104779","DOIUrl":"10.1016/j.cellimm.2023.104779","url":null,"abstract":"<div><p>Inflammatory bowel diseases are associated with dysregulated inflammatory immune responses in the gastrointestinal tract. We found that deficiencies of both IL-4 receptor alpha chain (IL-4Rα) and IL-10 in BALB/c mice (IL-4Rα × IL-10 KO mice) highly induced spontaneous rectal prolapse and diarrhea. These mice also exhibited severe colitis in their cecum and colon and marked elevation of serum proinflammatory cytokines including TNFα and IFNγ. These pathologies were transmittable with their cecal contents containing <em>Helicobacter</em> spp. Their mesenteric LN cells produced TNFα and IFNγ in response to soluble <em>H. hepaticus</em> antigens and high titers of <em>H. hepaticus</em>-specific serum IgG were also detected. These results suggested the important function of IL-4Rα signaling in controlling the intestinal inflammation and the susceptibility to intestinal microbes including <em>H. hepaticus</em>. Therefore, these IL-4Rα × IL-10 KO mice potentially provide the significant murine model for clarifying the causes and control of spontaneous colitis and intestinal inflammation.</p></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"393 ","pages":"Article 104779"},"PeriodicalIF":4.3,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71478443","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-11-01DOI: 10.1016/j.cellimm.2023.104783
Fangfang Liang , Cheng Peng , Xianze Luo , Linlin Wang , Yanyan Huang , Le Yin , Luming Yue , Jun Yang , Xiaodong Zhao
Wiskott-Aldrich syndrome (WAS) is a disorder characterized by rare X-linked genetic immune deficiency with mutations in the Was gene, which is specifically expressed in hematopoietic cells. The spleen plays a major role in hematopoiesis and red blood cell clearance. However, to date, comprehensive analyses of the spleen in wild-type (WT) and WASp-deficient (WAS-KO) mice, especially at the transcriptome level, have not been reported. In this study, single-cell RNA sequencing (scRNA-seq) was adopted to identify various types of immune cells and investigate the mechanisms underlying immune deficiency. We identified 30 clusters and 10 major cell subtypes among 11,269 cells; these cell types included B cells, T cells, dendritic cells (DCs), natural killer (NK) cells, monocytes, macrophages, granulocytes, stem cells and erythrocytes. Moreover, we evaluated gene expression differences among cell subtypes, identified differentially expressed genes (DEGs), and performed enrichment analyses to identify the reasons for the dysfunction in these different cell populations in WAS. Furthermore, some key genes were identified based on a comparison of the DEGs in each cell type involved in specific and nonspecific immune responses, and further analysis showed that these key genes were previously undiscovered pathology-related genes in WAS-KO mice. In summary, we present a landscape of immune cells in the spleen of WAS-KO mice based on detailed data obtained at single-cell resolution. These unprecedented data revealed the transcriptional characteristics of specific and nonspecific immune cells, and the key genes were identified, laying a foundation for future studies of WAS, especially studies into novel and underexplored mechanisms that may improve gene therapies for WAS.
{"title":"A single-cell atlas of immunocytes in the spleen of a mouse model of Wiskott-Aldrich syndrome","authors":"Fangfang Liang , Cheng Peng , Xianze Luo , Linlin Wang , Yanyan Huang , Le Yin , Luming Yue , Jun Yang , Xiaodong Zhao","doi":"10.1016/j.cellimm.2023.104783","DOIUrl":"10.1016/j.cellimm.2023.104783","url":null,"abstract":"<div><p>Wiskott-Aldrich syndrome (WAS) is a disorder characterized by rare X-linked genetic immune deficiency with mutations in the <em>Was</em> gene, which is specifically expressed in hematopoietic cells. The spleen plays a major role in hematopoiesis and red blood cell clearance. However, to date, comprehensive analyses of the spleen in wild-type (WT) and WASp-deficient (WAS-KO) mice, especially at the transcriptome level, have not been reported. In this study, single-cell RNA sequencing (scRNA-seq) was adopted to identify various types of immune cells and investigate the mechanisms underlying immune deficiency. We identified 30 clusters and 10 major cell subtypes among 11,269 cells; these cell types included B cells, T cells, dendritic cells (DCs), natural killer (NK) cells, monocytes, macrophages, granulocytes, stem cells and erythrocytes. Moreover, we evaluated gene expression differences among cell subtypes, identified differentially expressed genes (DEGs), and performed enrichment analyses to identify the reasons for the dysfunction in these different cell populations in WAS. Furthermore, some key genes were identified based on a comparison of the DEGs in each cell type involved in specific and nonspecific immune responses, and further analysis showed that these key genes were previously undiscovered pathology-related genes in WAS-KO mice. In summary, we present a landscape of immune cells in the spleen of WAS-KO mice based on detailed data obtained at single-cell resolution. These unprecedented data revealed the transcriptional characteristics of specific and nonspecific immune cells, and the key genes were identified, laying a foundation for future studies of WAS, especially studies into novel and underexplored mechanisms that may improve gene therapies for WAS.</p></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"393 ","pages":"Article 104783"},"PeriodicalIF":4.3,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"72013720","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-11-01DOI: 10.1016/j.cellimm.2023.104780
Brenal K. Singh , Yuichi Yokoyama , Yukinori Tanaka , Dorottya Laczkó , Deepak A. Deshpande , Taku Kambayashi
Allergic airway diseases are caused by inappropriate immune responses directed against inhaled environmental antigens. We previously reported that the inhibition of diacylglycerol (DAG) kinase ζ (DGKζ), an enzyme that terminates DAG-mediated signaling, protects against T cell-mediated allergic airway inflammation by blocking Th2 cell differentiation. In this study, we tested whether DGKζ deficiency also affects allergic airway disease mediated by type 2 innate lymphoid cells (ILC2)s. DGKζ-deficient mice displayed diminished ILC2 function and reduced papain-induced airway inflammation compared to wildtype mice. Unexpectedly, however, mice with hematopoietic cell-specific deletion of DGKζ displayed intact airway inflammation upon papain challenge. Rather, bone marrow chimera studies revealed that DGKζ deficiency in the non-hematopoietic compartment was responsible for the reduction in papain-induced airway inflammation. These data suggest that DGK might represent a novel therapeutic target not only for T cell-dependent but also ILC2-dependent allergic airway inflammation by affecting non-hematopoietic cells.
{"title":"Diacylglycerol kinase zeta deficiency attenuates papain-induced type 2 airway inflammation","authors":"Brenal K. Singh , Yuichi Yokoyama , Yukinori Tanaka , Dorottya Laczkó , Deepak A. Deshpande , Taku Kambayashi","doi":"10.1016/j.cellimm.2023.104780","DOIUrl":"10.1016/j.cellimm.2023.104780","url":null,"abstract":"<div><p>Allergic airway diseases are caused by inappropriate immune responses directed against inhaled environmental antigens. We previously reported that the inhibition of diacylglycerol (DAG) kinase<!--> <!-->ζ (DGKζ),<!--> <!-->an enzyme that terminates DAG-mediated signaling,<!--> <!-->protects against T cell-mediated allergic airway inflammation by blocking Th2 cell differentiation.<!--> <!-->In this study, we tested whether DGKζ deficiency also affects allergic airway disease mediated by type 2 innate lymphoid cells (ILC2)s. DGKζ-deficient mice displayed diminished ILC2 function and reduced papain-induced airway inflammation compared to wildtype mice. Unexpectedly, however, mice with hematopoietic cell-specific deletion of<!--> <span>DGKζ displayed intact airway inflammation upon papain challenge. Rather, bone marrow chimera studies revealed that</span> <!-->DGKζ deficiency in the non-hematopoietic compartment was responsible for the reduction in papain-induced airway inflammation. These data suggest that DGK might represent a novel therapeutic target not only for T cell-dependent but also ILC2-dependent allergic airway inflammation by affecting non-hematopoietic cells.</p></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"393 ","pages":"Article 104780"},"PeriodicalIF":4.3,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71421054","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-11-01DOI: 10.1016/j.cellimm.2023.104786
Naina Monga , Shikha Sharma , Ruchika Bhatia , Mahendra Bishnoi , Kanthi Kiran Kondepudi , Amarjit S. Naura
Given the reported role of gut-microbiota in asthma pathogenesis, the present work was carried to evaluate immunomodulatory action of newly isolated lactic acid producing bacterial strains Bifidobacterium breve Bif11 and Lactiplantibacillus plantarum LAB31 against asthma using ovalbumin (OVA) based mouse model. Our results show that both strains modulate Th2 immune response potentially through production of short chain fatty acids (SCFAs), resulting in suppression of OVA-induced airway inflammation. Furthermore, synbiotic comprising of both strains and prebiotic, Isomaltooligosaccharide exhibited superior potential in amelioration of OVA-induced airway inflammation through improved modulation of Th2 immune response. Further, synbiotic protects against OVA-induced mucus hyper-production and airway-hyperresponsiveness. Such protection was associated with normalization of gut microbiome and enhanced production of SCFAs in cecum which correlates closely with population of T-regulatory cells in spleen. Overall, our novel synbiotic possesses the ability to fine-tune the immune response for providing protection against allergic asthma.
{"title":"Immunomodulatory action of synbiotic comprising of newly isolated lactic acid producing bacterial strains against allergic asthma in mice","authors":"Naina Monga , Shikha Sharma , Ruchika Bhatia , Mahendra Bishnoi , Kanthi Kiran Kondepudi , Amarjit S. Naura","doi":"10.1016/j.cellimm.2023.104786","DOIUrl":"10.1016/j.cellimm.2023.104786","url":null,"abstract":"<div><p>Given the reported role of gut-microbiota in asthma pathogenesis, the present work was carried to evaluate immunomodulatory action of newly isolated lactic acid producing bacterial strains <em>Bifidobacterium breve</em> Bif11 and <em>Lactiplantibacillus plantarum</em> LAB31 against asthma using ovalbumin (OVA) based mouse model. Our results show that both strains modulate Th2 immune response potentially through production of short chain fatty acids (SCFAs), resulting in suppression of OVA-induced airway inflammation. Furthermore, synbiotic comprising of both strains and prebiotic, Isomaltooligosaccharide exhibited superior potential in amelioration of OVA-induced airway inflammation through improved modulation of Th2 immune response. Further, synbiotic protects against OVA-induced mucus hyper-production and airway-hyperresponsiveness. Such protection was associated with normalization of gut microbiome and enhanced production of SCFAs in cecum which correlates closely with population of T-regulatory cells in spleen. Overall, our novel synbiotic possesses the ability to fine-tune the immune response for providing protection against allergic asthma.</p></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"393 ","pages":"Article 104786"},"PeriodicalIF":4.3,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"135615316","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The administration of blinatumomab was accompanied by several adverse effects, including activation of regulatory T-cells and cytokine storm. The objective of this study was to produce and evaluate a novel αCD8/CD19 BiTE (αCD8/CD19) with the potency to directly target CD8+T-cells. In-silico studies were utilized for determining proper folding, receptor binding, and structural stability of αCD8/CD19 protein. Western blotting and indirect surface staining were used to evaluate the size accuracy and binding potency of the purified protein. Functionality was assessed for granzyme B production, cytotoxicity, and proliferation. The αCD8/CD19 recombinant protein was produced in the CHO-K1 cell line with a final concentration of 1.94 mg/l. The αCD8/CD19 bound to CD8+and CD19+ cell lines and induced significant granzyme B production, cytotoxic activity and proliferation potential in the presence of IL-2 and tumor target cells. The maximum CD8+T-cell biological activity was observed on the 10th day with 10:1 effector-to-target ratio.
{"title":"Anti-CD19/CD8 bispecific T cell engager for the potential treatment of B cell malignancies","authors":"Nafiseh Maghsoodi , Mohammadrasul Zareinejad , Ali Golestan , Elham Mahmoudi Maymand , Amin Ramezani","doi":"10.1016/j.cellimm.2023.104787","DOIUrl":"https://doi.org/10.1016/j.cellimm.2023.104787","url":null,"abstract":"<div><p>The administration of blinatumomab was accompanied by several adverse effects, including activation of regulatory <em>T</em>-cells and cytokine storm. The objective of this study was to produce and evaluate a novel αCD8/CD19 BiTE (αCD8/CD19) with the potency to directly target CD8<sup>+</sup> <em>T</em>-cells. In-silico studies were utilized for determining proper folding, receptor binding, and structural stability of αCD8/CD19 protein. Western blotting and indirect surface staining were used to evaluate the size accuracy and binding potency of the purified protein. Functionality was assessed for granzyme B production, cytotoxicity, and proliferation. The<!--> <!-->αCD8/CD19<!--> <!-->recombinant protein was produced in the CHO-K1 cell line with a final concentration of 1.94 mg/l. The αCD8/CD19 bound to CD8<sup>+</sup> <span>and CD19</span><sup>+</sup> <!-->cell lines and induced significant granzyme B production, cytotoxic activity and proliferation potential in the presence of IL-2 and tumor target cells. The maximum CD8<sup>+</sup> <em>T</em>-cell biological activity was observed on the 10th day with 10:1 effector-to-target ratio.</p></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"393 ","pages":"Article 104787"},"PeriodicalIF":4.3,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"134656940","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ro52 (TRIM21) belongs to the ubiquitin ligase family. This protein plays a crucial role in many immunological processes, including antibody-dependent intracellular neutralization, synergy with the complement system, antiviral response, death mediation, oxidative stress response, and protein ubiquitination. Abnormal expression of TRIM21 can break immunological tolerance and lead to the production of autoantibodies against TRIM21. Antibodies against TRIM21 are detected in various autoimmune diseases, including Sjögren's syndrome (SS), systemic lupus erythematosus (SLE), or myositis. However, anti-TRIM21 presence is not limited to autoimmune connective tissue disorders. It was observed in patients with malignancies, various cancerous processes, infectious diseases, and idiopathic interstitial pneumonia. The occurrence of TRIM21 autoantibodies is also associated with clinical features, such as the prevalence of interstitial lung diseases and cardiac or haematological involvement in connective tissue disorders. The purpose of this review was to summarize current knowledge of the immunological functions of TRIM21 and analyze the clinical implications of anti-TRIM21 antibodies in the disease course.
{"title":"Ro52/TRIM21 – From host defense to autoimmunity","authors":"Emilia Holwek , Aleksandra Opinc-Rosiak , Joanna Sarnik , Joanna Makowska","doi":"10.1016/j.cellimm.2023.104776","DOIUrl":"10.1016/j.cellimm.2023.104776","url":null,"abstract":"<div><p>Ro52 (TRIM21) belongs to the ubiquitin ligase family. This protein plays a crucial role in many immunological processes, including antibody-dependent intracellular neutralization, synergy with the complement system, antiviral response, death mediation, oxidative stress response, and protein ubiquitination. Abnormal expression of TRIM21 can break immunological tolerance and lead to the production of autoantibodies against TRIM21. Antibodies against TRIM21 are detected in various autoimmune diseases, including Sjögren's syndrome (SS), systemic lupus erythematosus (SLE), or myositis. However, anti-TRIM21 presence is not limited to autoimmune connective tissue disorders. It was observed in patients with malignancies, various cancerous processes, infectious diseases, and idiopathic interstitial pneumonia. The occurrence of TRIM21 autoantibodies is also associated with clinical features, such as the prevalence of interstitial lung diseases and cardiac or haematological involvement in connective tissue disorders. The purpose of this review was to summarize current knowledge of the immunological functions of TRIM21 and analyze the clinical implications of anti-TRIM21 antibodies in the disease course.</p></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"393 ","pages":"Article 104776"},"PeriodicalIF":4.3,"publicationDate":"2023-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49674773","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}