Adrian Ys Lee, Zhankun Qi, Katherine Jl Jackson, Joanne H Reed
Sjögren's disease (SjD) is a chronic autoimmune disorder characterized by increased circulating self-reactive B cells. While many of these self-reactive B cells emerge from the bone marrow, it is not known whether they are excluded from or enriched in specific developmental stages in the periphery. The aim of this study was to determine the immunophenotype of circulating self-reactive B cells in SjD to inform more precise therapeutic targeting. Five major B cell populations: transitional, mature naïve, switched memory, double negative and plasmablasts were single-cell sorted and cultured to produce IgG. Self-reactive IgG was identified by ELISA, flow cytometry of permeabilized HEK293 cells and HEp-2 indirect immunofluorescence. Immunoglobulin heavy chains were sequenced by Sanger and next-generation sequencing. Compared with healthy donor controls (HCs), SjD patients had higher frequencies of naïve and CD21low atypical memory B cell subsets, while antigen-experienced B cells expressed more Ki67 and CD86. B cells recognizing intracellular self-antigens were identified in all stages of peripheral B cell development for SjD and HCs, but frequencies of autoreactive B cells were up to 10-fold higher in SjD. Self-reactive transitional B cells expressed higher surface CD38 and lower surface IgM. An increase in self-reactive B cells throughout peripheral development in SjD compared with HCs suggests that counterselection of autoantibody-bearing B cells during central and peripheral tolerance checkpoints are reduced in SjD. Therapeutic strategies focused on depleting B cells based on B cell receptor specificity rather than the developmental stage would be more efficient to target self-reactive B cells in SjD.
{"title":"Self-reactive B cells are increased in all major stages of peripheral development in Sjögren's disease.","authors":"Adrian Ys Lee, Zhankun Qi, Katherine Jl Jackson, Joanne H Reed","doi":"10.1111/imcb.70005","DOIUrl":"https://doi.org/10.1111/imcb.70005","url":null,"abstract":"<p><p>Sjögren's disease (SjD) is a chronic autoimmune disorder characterized by increased circulating self-reactive B cells. While many of these self-reactive B cells emerge from the bone marrow, it is not known whether they are excluded from or enriched in specific developmental stages in the periphery. The aim of this study was to determine the immunophenotype of circulating self-reactive B cells in SjD to inform more precise therapeutic targeting. Five major B cell populations: transitional, mature naïve, switched memory, double negative and plasmablasts were single-cell sorted and cultured to produce IgG. Self-reactive IgG was identified by ELISA, flow cytometry of permeabilized HEK293 cells and HEp-2 indirect immunofluorescence. Immunoglobulin heavy chains were sequenced by Sanger and next-generation sequencing. Compared with healthy donor controls (HCs), SjD patients had higher frequencies of naïve and CD21<sup>low</sup> atypical memory B cell subsets, while antigen-experienced B cells expressed more Ki67 and CD86. B cells recognizing intracellular self-antigens were identified in all stages of peripheral B cell development for SjD and HCs, but frequencies of autoreactive B cells were up to 10-fold higher in SjD. Self-reactive transitional B cells expressed higher surface CD38 and lower surface IgM. An increase in self-reactive B cells throughout peripheral development in SjD compared with HCs suggests that counterselection of autoantibody-bearing B cells during central and peripheral tolerance checkpoints are reduced in SjD. Therapeutic strategies focused on depleting B cells based on B cell receptor specificity rather than the developmental stage would be more efficient to target self-reactive B cells in SjD.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143432001","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}
Rhea J Longley, Andre L Samson, Georgia Atkin-Smith, Alex R Carey Hulyer, Trent Ashton, Nadia M Davidson
In this article, we discuss our experiences and perspectives in forming a workplace Parents Group. We reflect on the need for these networks, what has worked well, and the challenges we've experienced. We also provide some practical advice for those with parenting-related career disruptions for addressing this topic in grant applications.
{"title":"From pipettes to playdates: establishing a parent support group in a research setting.","authors":"Rhea J Longley, Andre L Samson, Georgia Atkin-Smith, Alex R Carey Hulyer, Trent Ashton, Nadia M Davidson","doi":"10.1111/imcb.70004","DOIUrl":"https://doi.org/10.1111/imcb.70004","url":null,"abstract":"<p><p>In this article, we discuss our experiences and perspectives in forming a workplace Parents Group. We reflect on the need for these networks, what has worked well, and the challenges we've experienced. We also provide some practical advice for those with parenting-related career disruptions for addressing this topic in grant applications.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143431994","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}
We recently developed a hybrid protein, tentatively named human MIKO-1 (hMIKO-1), based on the amino acid sequences of human S100A8 (hS100A8) and hS100A9. Human THP-1 macrophages (THP-1m), differentiated from THP-1 cells by phorbol 12-myristate 13-acetate, were used to investigate the immune function of hMIKO-1 as a drug for inflammatory diseases. Western blotting was conducted to confirm whether hMIKO-1 binds with β-actin and nuclear factor-kappa B to form complexes in THP-1m. A polymerase chain reaction (PCR) and quantitative PCR were performed to examine changes in the messenger RNA levels of proinflammatory cytokines in THP-1m. Fluorescent immunochemical staining was used to observe the intracellular localization of hMIKO-1 and hS100A8 or hS100A9 in THP-1m. As observed microscopically, the intracellular localization of hMIKO-1 in THP-1m was consistent with that of hS100A8, suggesting the close involvement of hS100A8 in the intracellular behavior of hMIKO-1 in THP-1m. Western blotting revealed that hMIKO-1 formed complexes with intracellular proteins, such as β-actin and nuclear factor-kappa B, to negatively regulate inflammatory signal transduction in THP-1m. Flow cytometry showed that the binding of hMIKO-1 to THP-1m significantly decreased when THP-1m were preliminarily treated with a sialidase (neuraminidases) cocktail. Therefore, the present results strongly suggest that the binding of hMIKO-1 to THP-1m closely involves the sugar chains of the surface proteins of cells. The messenger RNA expression of each proinflammatory cytokine was significantly suppressed in THP-1m preliminarily treated with hMIKO-1 despite a subsequent stimulation with lipopolysaccharide. In conclusion, hMIKO-1 is a functional molecule that significantly inhibits inflammatory signal transduction in THP-1m.
{"title":"A hybrid protein is a functional molecule to reduce the cytokine storm caused by excessively activated macrophages.","authors":"Masaki Ikemoto, Takuya Kotani, Kohki Okada, Shogo Matsuda, Tohru Takeuchi","doi":"10.1111/imcb.70000","DOIUrl":"https://doi.org/10.1111/imcb.70000","url":null,"abstract":"<p><p>We recently developed a hybrid protein, tentatively named human MIKO-1 (hMIKO-1), based on the amino acid sequences of human S100A8 (hS100A8) and hS100A9. Human THP-1 macrophages (THP-1m), differentiated from THP-1 cells by phorbol 12-myristate 13-acetate, were used to investigate the immune function of hMIKO-1 as a drug for inflammatory diseases. Western blotting was conducted to confirm whether hMIKO-1 binds with β-actin and nuclear factor-kappa B to form complexes in THP-1m. A polymerase chain reaction (PCR) and quantitative PCR were performed to examine changes in the messenger RNA levels of proinflammatory cytokines in THP-1m. Fluorescent immunochemical staining was used to observe the intracellular localization of hMIKO-1 and hS100A8 or hS100A9 in THP-1m. As observed microscopically, the intracellular localization of hMIKO-1 in THP-1m was consistent with that of hS100A8, suggesting the close involvement of hS100A8 in the intracellular behavior of hMIKO-1 in THP-1m. Western blotting revealed that hMIKO-1 formed complexes with intracellular proteins, such as β-actin and nuclear factor-kappa B, to negatively regulate inflammatory signal transduction in THP-1m. Flow cytometry showed that the binding of hMIKO-1 to THP-1m significantly decreased when THP-1m were preliminarily treated with a sialidase (neuraminidases) cocktail. Therefore, the present results strongly suggest that the binding of hMIKO-1 to THP-1m closely involves the sugar chains of the surface proteins of cells. The messenger RNA expression of each proinflammatory cytokine was significantly suppressed in THP-1m preliminarily treated with hMIKO-1 despite a subsequent stimulation with lipopolysaccharide. In conclusion, hMIKO-1 is a functional molecule that significantly inhibits inflammatory signal transduction in THP-1m.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143424631","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}
Over the years, beginning with my undergraduate lab experiences, I became aware of the traits essential for success in academic study and research. The traits, which I refer to as the "S Triple G Traits" are Support, Growth mindset, Grit and Grace. Support can come from a person's family, friends, lab members, supervisors and themselves. A growth mindset is being open to the possibility of improving and overcoming obstacles. Grit can be considered the process of continuing to apply what you learn from reflection of having a growth mindset and setting it into practice. In theory, your growth mindset can be the foundation on which you can propel your grit in the right direction. Grace is taking a moment to acknowledge your setback and give yourself permission to honor and recognize all the effort you contributed. Using this combination of traits will help to equip students to achieve their goals when faced with obstacles.
{"title":"The S Triple G Traits - essential traits for students.","authors":"Jasmine G Hughes","doi":"10.1111/imcb.70001","DOIUrl":"https://doi.org/10.1111/imcb.70001","url":null,"abstract":"<p><p>Over the years, beginning with my undergraduate lab experiences, I became aware of the traits essential for success in academic study and research. The traits, which I refer to as the \"S Triple G Traits\" are Support, Growth mindset, Grit and Grace. Support can come from a person's family, friends, lab members, supervisors and themselves. A growth mindset is being open to the possibility of improving and overcoming obstacles. Grit can be considered the process of continuing to apply what you learn from reflection of having a growth mindset and setting it into practice. In theory, your growth mindset can be the foundation on which you can propel your grit in the right direction. Grace is taking a moment to acknowledge your setback and give yourself permission to honor and recognize all the effort you contributed. Using this combination of traits will help to equip students to achieve their goals when faced with obstacles.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143381353","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 glycolytic enzyme, fructose-1,6-bisphosphate aldolase B (ALDOB), is recognized for its key role in shaping tthe umor immune microenvironment. However, the precise ways in which it influences the CD8+ T cell immune response in colorectal cancer (CRC) are still largely unknown. This study is designed to elucidate the interplay between ALDOB and the immune system in CRC. We analyzed the high expression of ALDOB in CRC tissues and cells through bioinformatics, clinical samples and in vitro experiments, finding that it promoted tumor progression. Its high expression was negatively correlated with CD8 expression and positively correlated with PDL1 expression. Further cell experiments revealed that ALDOB overexpression enhanced the expression of WNT signaling pathway-related proteins (β-catenin and c-myc), which in turn promoted PDL1 expression in CRC cells, inhibiting the proliferation and killing effect of CD8+ T cells in co-culture systems. Our findings disclose how ALDOB influences CD8+ T cell recruitment and antitumor immune function, proposing it as a potential target for the treatment of CRC.
{"title":"ALDOB suppresses the activity of CD8<sup>+</sup> T cells in colorectal cancer via the WNT signaling pathway.","authors":"Jinwei Liu, Chao Hu, Yuan Jin","doi":"10.1111/imcb.12853","DOIUrl":"https://doi.org/10.1111/imcb.12853","url":null,"abstract":"<p><p>The glycolytic enzyme, fructose-1,6-bisphosphate aldolase B (ALDOB), is recognized for its key role in shaping tthe umor immune microenvironment. However, the precise ways in which it influences the CD8<sup>+</sup> T cell immune response in colorectal cancer (CRC) are still largely unknown. This study is designed to elucidate the interplay between ALDOB and the immune system in CRC. We analyzed the high expression of ALDOB in CRC tissues and cells through bioinformatics, clinical samples and in vitro experiments, finding that it promoted tumor progression. Its high expression was negatively correlated with CD8 expression and positively correlated with PDL1 expression. Further cell experiments revealed that ALDOB overexpression enhanced the expression of WNT signaling pathway-related proteins (β-catenin and c-myc), which in turn promoted PDL1 expression in CRC cells, inhibiting the proliferation and killing effect of CD8<sup>+</sup> T cells in co-culture systems. Our findings disclose how ALDOB influences CD8<sup>+</sup> T cell recruitment and antitumor immune function, proposing it as a potential target for the treatment of CRC.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143254258","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}
Jessica G Borger, Rhea J Longley, Megan F Taylor, Ruben Motrich, Jennifer Ae Payne, Roslyn A Kemp
The discourse surrounding gender equity has intensified recently, amplified by the impacts of the COVID-19 pandemic, highlighting the critical underrepresentation of women in leadership roles across various sectors including the media and healthcare. In medical research, this disparity is particularly pronounced, with women often excluded from senior positions despite their substantial presence in the workforce. This review seeks to explore the multifaceted issue of gender inequity in medical research leadership, examining the systemic barriers that women face, the socioeconomic factors that compound these challenges and the global variations in leadership representation of women. Diverse leadership teams are essential for fostering medical innovation, improving patient outcomes and ensuring that clinical trials and medical research are effective, inclusive and representative. The underrepresentation of women in leadership roles is not merely a matter of gender bias; it is intricately linked to socioeconomic factors that hinder their advancement. Women from lower socioeconomic backgrounds face additional obstacles, such as limited access to education and professional networks, which further exacerbate their underrepresentation in leadership positions. Moreover, cultural and societal norms play a significant role in shaping the career trajectories of women. As a group of immunologists, including representatives of the International Union of Immunological Sciences (IUIS) Gender Equity Committee, we review the causes of these inequities. We examine the impact of gender-diverse leadership on pre-clinical and medical research, emphasizing the need for inclusive leadership to drive progress in medical research and resulting healthcare. Finally, the review proposes strategies for improving gender equity in medical research leadership, including policy changes, organizational initiatives and societal shifts. By addressing these critical issues, this review contributes to the ongoing efforts to promote gender equity in medical research, ultimately enhancing the quality and inclusiveness of scientific inquiry and its impact on healthcare delivery.
{"title":"Global perspectives to enhance strategies for advancing women in healthcare and STEMM leadership.","authors":"Jessica G Borger, Rhea J Longley, Megan F Taylor, Ruben Motrich, Jennifer Ae Payne, Roslyn A Kemp","doi":"10.1111/imcb.12854","DOIUrl":"https://doi.org/10.1111/imcb.12854","url":null,"abstract":"<p><p>The discourse surrounding gender equity has intensified recently, amplified by the impacts of the COVID-19 pandemic, highlighting the critical underrepresentation of women in leadership roles across various sectors including the media and healthcare. In medical research, this disparity is particularly pronounced, with women often excluded from senior positions despite their substantial presence in the workforce. This review seeks to explore the multifaceted issue of gender inequity in medical research leadership, examining the systemic barriers that women face, the socioeconomic factors that compound these challenges and the global variations in leadership representation of women. Diverse leadership teams are essential for fostering medical innovation, improving patient outcomes and ensuring that clinical trials and medical research are effective, inclusive and representative. The underrepresentation of women in leadership roles is not merely a matter of gender bias; it is intricately linked to socioeconomic factors that hinder their advancement. Women from lower socioeconomic backgrounds face additional obstacles, such as limited access to education and professional networks, which further exacerbate their underrepresentation in leadership positions. Moreover, cultural and societal norms play a significant role in shaping the career trajectories of women. As a group of immunologists, including representatives of the International Union of Immunological Sciences (IUIS) Gender Equity Committee, we review the causes of these inequities. We examine the impact of gender-diverse leadership on pre-clinical and medical research, emphasizing the need for inclusive leadership to drive progress in medical research and resulting healthcare. Finally, the review proposes strategies for improving gender equity in medical research leadership, including policy changes, organizational initiatives and societal shifts. By addressing these critical issues, this review contributes to the ongoing efforts to promote gender equity in medical research, ultimately enhancing the quality and inclusiveness of scientific inquiry and its impact on healthcare delivery.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143187808","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}
In this article, we discuss a recent study, where autosomal monoallelic expression of genes underlying Inborn Errors of Immunity were investigated. About 2-10% of genes are predominantly transcribed from a single allele leading to autosomal random monoallelic expression (I). If this is skewed in a cell population from an individual with an autosomal dominant inborn error of immunity, this can lead to a mild to no phenotype (incomplete penetrance) if the wildtype allele is favored (II), or to more severe disease presentation if the variant allele is favored (III).
{"title":"A potential role for monoallelic expression in penetrance of autosomal dominant inborn errors of immunity.","authors":"Emily Sj Edwards, Menno C van Zelm","doi":"10.1111/imcb.12856","DOIUrl":"https://doi.org/10.1111/imcb.12856","url":null,"abstract":"<p><p>In this article, we discuss a recent study, where autosomal monoallelic expression of genes underlying Inborn Errors of Immunity were investigated. About 2-10% of genes are predominantly transcribed from a single allele leading to autosomal random monoallelic expression (I). If this is skewed in a cell population from an individual with an autosomal dominant inborn error of immunity, this can lead to a mild to no phenotype (incomplete penetrance) if the wildtype allele is favored (II), or to more severe disease presentation if the variant allele is favored (III).</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143254250","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}
Indrani Nayak, Rosalba Biondo, William C Stewart, Rebecca J Fulton, Nina Möker, Congcong Zhang, Salim I Khakoo, Jayajit Das
Natural killer (NK) cells are emerging agents for cancer therapy. Several different cytokines are used to generate NK cells for adoptive immunotherapy including interleukin (IL)-2, IL-12, IL-15 and IL-18 in solution, and membrane-bound IL-21. These cytokines drive NK cell activation through the integration of signal transducers and activators of transcription (STAT) and nuclear factor-kappa B (NF-κB) pathways, which overlap and synergize, making it challenging to predict optimal cytokine combinations for both proliferation and cytotoxicity. We integrated functional assays for NK cells cultured in a variety of cytokine combinations with mathematical modeling using feature selection and mechanistic regression models. Our regression model successfully predicts NK cell proliferation for different cytokine combinations and indicates synergy of activated STATs and NF-κB transcription factors between priming and post-priming phases. The use of IL-21 in solution in the priming of NK cell culture resulted in an improved NK cell proliferation, without compromising cytotoxicity potential or interferon gamma secretion against hepatocellular carcinoma cell lines. Our work provides an integrative framework for interrogating NK cell proliferation and activation for cancer immunotherapy.
自然杀伤(NK)细胞是新兴的癌症治疗药物。有几种不同的细胞因子可用于产生NK细胞,用于采纳性免疫疗法,包括溶液中的白细胞介素(IL)-2、IL-12、IL-15和IL-18,以及膜结合的IL-21。这些细胞因子通过信号转导和转录激活因子(STAT)与核因子-卡巴B(NF-κB)途径的整合来驱动NK细胞的活化,这两种途径会重叠并产生协同作用,因此预测细胞因子的最佳增殖和细胞毒性组合具有挑战性。我们将多种细胞因子组合培养的 NK 细胞的功能测试与使用特征选择和机理回归模型的数学建模相结合。我们的回归模型成功预测了不同细胞因子组合的 NK 细胞增殖情况,并显示了激活的 STAT 和 NF-κB 转录因子在引物阶段和引物后阶段之间的协同作用。在 NK 细胞培养的启动阶段使用溶液中的 IL-21 可改善 NK 细胞的增殖,同时不影响细胞毒性潜力或针对肝细胞癌细胞系的伽马干扰素分泌。我们的工作提供了一个综合框架,可用于研究癌症免疫疗法中的 NK 细胞增殖和活化。
{"title":"Modeling the response to interleukin-21 to inform natural killer cell immunotherapy","authors":"Indrani Nayak, Rosalba Biondo, William C Stewart, Rebecca J Fulton, Nina Möker, Congcong Zhang, Salim I Khakoo, Jayajit Das","doi":"10.1111/imcb.12848","DOIUrl":"10.1111/imcb.12848","url":null,"abstract":"<p>Natural killer (NK) cells are emerging agents for cancer therapy. Several different cytokines are used to generate NK cells for adoptive immunotherapy including interleukin (IL)-2, IL-12, IL-15 and IL-18 in solution, and membrane-bound IL-21. These cytokines drive NK cell activation through the integration of signal transducers and activators of transcription (STAT) and nuclear factor-kappa B (NF-κB) pathways, which overlap and synergize, making it challenging to predict optimal cytokine combinations for both proliferation and cytotoxicity. We integrated functional assays for NK cells cultured in a variety of cytokine combinations with mathematical modeling using feature selection and mechanistic regression models. Our regression model successfully predicts NK cell proliferation for different cytokine combinations and indicates synergy of activated STATs and NF-κB transcription factors between priming and post-priming phases. The use of IL-21 in solution in the priming of NK cell culture resulted in an improved NK cell proliferation, without compromising cytotoxicity potential or interferon gamma secretion against hepatocellular carcinoma cell lines. Our work provides an integrative framework for interrogating NK cell proliferation and activation for cancer immunotherapy.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":"103 2","pages":"192-212"},"PeriodicalIF":3.2,"publicationDate":"2025-01-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/imcb.12848","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143045129","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kaja Karaś, Joanna Pastwińska, Anna Sałkowska, Iwona Karwaciak, Rafał A Bachorz, Marcin Ratajewski
AT7519, which inhibits multiple cyclin-dependent kinases, has been extensively investigated in various types of cancer cells. Previous studies have demonstrated the ability of this molecule to suppress the expression of the nuclear receptor retinoic acid-related orphan receptor gamma (RORγ) and several genes involved in hepatocellular carcinoma progression. In this study, we identified a distinct agonistic effect of AT7519 on RORγt, an isoform expressed by various immune cells, including T helper 17 lymphocytes. These immune cells play pivotal roles in shaping the tumor microenvironment and promoting the anticancer response of the immune system. After exposure to AT7519 during differentiation, primary human CD4+ T cells presented increased expression of IL17A/F, IFNG and GZMB and decreased expression of PDCD1 and CTLA4. These findings elucidate a previously unrecognized facet of AT7519 activity and suggest the potential incorporation of this molecule into immune therapies to augment the effectiveness of diverse anticancer strategies involving anti-programmed cell death protein 1 (anti-PD-1) and anti-cytotoxic T-lymphocyte antigen 4 (anti-CTLA4) regimens.
{"title":"The cyclin-dependent kinase inhibitor AT7519 is a human RORγt agonist.","authors":"Kaja Karaś, Joanna Pastwińska, Anna Sałkowska, Iwona Karwaciak, Rafał A Bachorz, Marcin Ratajewski","doi":"10.1111/imcb.12851","DOIUrl":"https://doi.org/10.1111/imcb.12851","url":null,"abstract":"<p><p>AT7519, which inhibits multiple cyclin-dependent kinases, has been extensively investigated in various types of cancer cells. Previous studies have demonstrated the ability of this molecule to suppress the expression of the nuclear receptor retinoic acid-related orphan receptor gamma (RORγ) and several genes involved in hepatocellular carcinoma progression. In this study, we identified a distinct agonistic effect of AT7519 on RORγt, an isoform expressed by various immune cells, including T helper 17 lymphocytes. These immune cells play pivotal roles in shaping the tumor microenvironment and promoting the anticancer response of the immune system. After exposure to AT7519 during differentiation, primary human CD4<sup>+</sup> T cells presented increased expression of IL17A/F, IFNG and GZMB and decreased expression of PDCD1 and CTLA4. These findings elucidate a previously unrecognized facet of AT7519 activity and suggest the potential incorporation of this molecule into immune therapies to augment the effectiveness of diverse anticancer strategies involving anti-programmed cell death protein 1 (anti-PD-1) and anti-cytotoxic T-lymphocyte antigen 4 (anti-CTLA4) regimens.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031684","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}
Correction to: Immunology & Cell Biology 2024; https://doi.10.1111/imcb.12826
The name of one of the authors is incorrect. Ning Ming should be Ning Meng. The correct spelling of this author's name appears in the title above.
{"title":"ADAM10 modulates the efficacy of T-cell mediated therapy in solid tumors","authors":"Ahmed ME Abdalla, Yu Miao, Ning Meng, Chenxi Ouyang","doi":"10.1111/imcb.12855","DOIUrl":"10.1111/imcb.12855","url":null,"abstract":"<p><i>Immunology & Cell Biology</i> 2025; <b>103</b>: 213; https://doi.org/10.1111/imcb.12855</p><p>Correction to: <i>Immunology & Cell Biology</i> 2024; https://doi.10.1111/imcb.12826</p><p>The name of one of the authors is incorrect. Ning Ming should be Ning Meng. The correct spelling of this author's name appears in the title above.</p>","PeriodicalId":179,"journal":{"name":"Immunology & Cell Biology","volume":"103 2","pages":"213"},"PeriodicalIF":3.2,"publicationDate":"2025-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/imcb.12855","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142997009","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}