79 results on '"Pipkin, Matthew E."'
Search Results
52. Runx3 programs CD8+ T cell residency in non-lymphoid tissues and tumours.
- Author
-
Milner, J. Justin, Toma, Clara, Yu, Bingfei, Zhang, Kai, Omilusik, Kyla, Phan, Anthony T., Wang, Dapeng, Getzler, Adam J., Nguyen, Toan, Crotty, Shane, Wang, Wei, Pipkin, Matthew E., and Goldrath, Ananda W.
- Abstract
Tissue-resident memory CD8
+ T (TRM ) cells are found at common sites of pathogen exposure, where they elicit rapid and robust protective immune responses. However, the molecular signals that control TRM cell differentiation and homeostasis are not fully understood. Here we show that mouse TRM precursor cells represent a unique CD8+ T cell subset that is distinct from the precursors of circulating memory cell populations at the levels of gene expression and chromatin accessibility. Using computational and pooled in vivo RNA interference screens, we identify the transcription factor Runx3 as a key regulator of TRM cell differentiation and homeostasis. Runx3 was required to establish TRM cell populations in diverse tissue environments, and supported the expression of crucial tissue-residency genes while suppressing genes associated with tissue egress and recirculation. Furthermore, we show that human and mouse tumour-infiltrating lymphocytes share a core tissue-residency gene-expression signature with TRM cells that is associated with Runx3 activity. In a mouse model of adoptive T cell therapy for melanoma, Runx3-deficient CD8+ tumour-infiltrating lymphocytes failed to accumulate in tumours, resulting in greater rates of tumour growth and mortality. Conversely, overexpression of Runx3 enhanced tumour-specific CD8+ T cell abundance, delayed tumour growth, and prolonged survival. In addition to establishing Runx3 as a central regulator of TRM cell differentiation, these results provide insight into the signals that promote T cell residency in non-lymphoid sites, which could be used to enhance vaccine efficacy or adoptive cell therapy treatments that target cancer. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
53. The Transcription Factor NFAT Promotes Exhaustion of Activated CD8 + T Cells
- Author
-
Martinez, Gustavo J., primary, Pereira, Renata M., additional, Äijö, Tarmo, additional, Kim, Edward Y., additional, Marangoni, Francesco, additional, Pipkin, Matthew E., additional, Togher, Susan, additional, Heissmeyer, Vigo, additional, Zhang, Yi Chen, additional, Crotty, Shane, additional, Lamperti, Edward D., additional, Ansel, K. Mark, additional, Mempel, Thorsten R., additional, Lähdesmäki, Harri, additional, Hogan, Patrick G., additional, and Rao, Anjana, additional
- Published
- 2015
- Full Text
- View/download PDF
54. Runx3 programs CD8+T cell residency in non-lymphoid tissues and tumours
- Author
-
Milner, J. Justin, Toma, Clara, Yu, Bingfei, Zhang, Kai, Omilusik, Kyla, Phan, Anthony T., Wang, Dapeng, Getzler, Adam J., Nguyen, Toan, Crotty, Shane, Wang, Wei, Pipkin, Matthew E., and Goldrath, Ananda W.
- Abstract
Tissue-resident memory CD8+T (TRM) cells are found at common sites of pathogen exposure, where they elicit rapid and robust protective immune responses. However, the molecular signals that control TRMcell differentiation and homeostasis are not fully understood. Here we show that mouse TRMprecursor cells represent a unique CD8+T cell subset that is distinct from the precursors of circulating memory cell populations at the levels of gene expression and chromatin accessibility. Using computational and pooled in vivo RNA interference screens, we identify the transcription factor Runx3 as a key regulator of TRMcell differentiation and homeostasis. Runx3 was required to establish TRMcell populations in diverse tissue environments, and supported the expression of crucial tissue-residency genes while suppressing genes associated with tissue egress and recirculation. Furthermore, we show that human and mouse tumour-infiltrating lymphocytes share a core tissue-residency gene-expression signature with TRMcells that is associated with Runx3 activity. In a mouse model of adoptive T cell therapy for melanoma, Runx3-deficient CD8+tumour-infiltrating lymphocytes failed to accumulate in tumours, resulting in greater rates of tumour growth and mortality. Conversely, overexpression of Runx3 enhanced tumour-specific CD8+T cell abundance, delayed tumour growth, and prolonged survival. In addition to establishing Runx3 as a central regulator of TRMcell differentiation, these results provide insight into the signals that promote T cell residency in non-lymphoid sites, which could be used to enhance vaccine efficacy or adoptive cell therapy treatments that target cancer.
- Published
- 2017
- Full Text
- View/download PDF
55. Epigenetic landscapes reveal transcription factors that regulate CD8+T cell differentiation
- Author
-
Yu, Bingfei, Zhang, Kai, Milner, J Justin, Toma, Clara, Chen, Runqiang, Scott-Browne, James P, Pereira, Renata M, Crotty, Shane, Chang, John T, Pipkin, Matthew E, Wang, Wei, and Goldrath, Ananda W
- Abstract
Dynamic changes in the expression of transcription factors (TFs) can influence the specification of distinct CD8+T cell fates, but the observation of equivalent expression of TFs among differentially fated precursor cells suggests additional underlying mechanisms. Here we profiled the genome-wide histone modifications, open chromatin and gene expression of naive, terminal-effector, memory-precursor and memory CD8+T cell populations induced during the in vivo response to bacterial infection. Integration of these data suggested that the expression and binding of TFs contributed to the establishment of subset-specific enhancers during differentiation. We developed a new bioinformatics method using the PageRank algorithm to reveal key TFs that influence the generation of effector and memory populations. The TFs YY1 and Nr3c1, both constitutively expressed during CD8+T cell differentiation, regulated the formation of terminal-effector cell fates and memory-precursor cell fates, respectively. Our data define the epigenetic landscape of differentiation intermediates and facilitate the identification of TFs with previously unappreciated roles in CD8+T cell differentiation.
- Published
- 2017
- Full Text
- View/download PDF
56. Memories in the snow: immune memory, persistent infection and chronic disease
- Author
-
Pipkin, Matthew E, primary
- Published
- 2011
- Full Text
- View/download PDF
57. SnapShot: Effector and Memory T Cell Differentiation
- Author
-
Pipkin, Matthew E., primary and Rao, Anjana, additional
- Published
- 2009
- Full Text
- View/download PDF
58. Runx3 and T-box proteins cooperate to establish the transcriptional program of effector CTLs
- Author
-
Cruz-Guilloty, Fernando, primary, Pipkin, Matthew E., additional, Djuretic, Ivana M., additional, Levanon, Ditsa, additional, Lotem, Joseph, additional, Lichtenheld, Mathias G., additional, Groner, Yoram, additional, and Rao, Anjana, additional
- Published
- 2009
- Full Text
- View/download PDF
59. MicroRNA-221–222 Regulate the Cell Cycle in Mast Cells
- Author
-
Mayoral, Ramon J., primary, Pipkin, Matthew E., additional, Pachkov, Mikhail, additional, van Nimwegen, Erik, additional, Rao, Anjana, additional, and Monticelli, Silvia, additional
- Published
- 2009
- Full Text
- View/download PDF
60. New Sequence of Strategies to Define Gene Transcriptional Territories in Lymphocytes: The Perforin Example (35.32)
- Author
-
Lichtenheld, Mathias Georg, primary, Zúñiga-Pflücker, Juan Carlos, additional, Rao, Anjana, additional, and Pipkin, Matthew E., additional
- Published
- 2007
- Full Text
- View/download PDF
61. Cbx3/HP1γ deficiency confers enhanced tumor-killing capacity on CD8+ T cells
- Author
-
Sun, Michael, Ha, Ngoc, Pham, Duc-Hung, Frederick, Megan, Sharma, Bandana, Naruse, Chie, Asano, Masahide, Pipkin, Matthew E., George, Rani E., and Thai, To-Ha
- Abstract
Cbx3/HP1γ is a histone reader whose function in the immune system is not completely understood. Here, we demonstrate that in CD8+ T cells, Cbx3/HP1γ insufficiency leads to chromatin remodeling accompanied by enhanced Prf1, Gzmb and Ifng expression. In tumors obtained from Cbx3/HP1γ-insufficient mice or wild type mice treated with Cbx3/HP1γ-insufficient CD8+ T cells, there is an increase of CD8+ effector T cells expressing the stimulatory receptor Klrk1/NKG2D, a decrease in CD4+ CD25+ FOXP3+ regulatory T cells (Treg cells) as well as CD25+ CD4+ T cells expressing the inhibitory receptor CTLA4. Together these changes in the tumor immune environment may have mitigated tumor burden in Cbx3/HP1γ-insufficient mice or wild type mice treated with Cbx3/HP1γ-insufficient CD8+ T cells. These findings suggest that targeting Cbx3/HP1γ can represent a rational therapeutic approach to control growth of solid tumors.
- Published
- 2017
- Full Text
- View/download PDF
62. MicroRNA-directed program of cytotoxic CD8+ T-cell differentiation.
- Author
-
Trifari, Sara, Pipkin, Matthew E., Bandukwala, Hozefa S., Äijö, Tarmo, Bassein, Jed, Runqiang Chen, Martinez, Gustavo J., and Rao, Anjana
- Subjects
- *
MICRORNA , *CD8 antigen , *T cells , *CELL differentiation , *INTERLEUKIN-2 - Abstract
Acquisition of effector properties is a key step in the generation of cytotoxic T lymphocytes (CTLs). Here we show that inflammatory signals regulate Dicer expression in CTLs, and that deletion or depletion of Dicer in mouse or human activated CD8+ T cells causes up-regulation of perforin, granzymes, and effector cytokines. Genome-wide analysis of microRNA (miR, miRNA) changes induced by exposure of differentiating CTLs to IL-2 and inflammatory signals identifies miR-139 and miR-150 as components of an miRNA network that controls perforin, eomesodermin, and IL-2Ra expression in differentiating CTLs and whose activity is modulated by IL-2, inflammation, and antigenic stimulation. Overall, our data show that strong IL-2R and inflammatory signals act through Dicer and miRNAs to control the cytolytic program and other aspects of effector CTL differentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
63. Erratum: Runx3 programs CD8+ T cell residency in non-lymphoid tissues and tumours.
- Author
-
Milner, J. Justin, Toma, Clara, Yu, Bingfei, Zhang, Kai, Omilusik, Kyla, Phan, Anthony T., Wang, Dapeng, Getzler, Adam J., Nguyen, Toan, Crotty, Shane, Wang, Wei, Pipkin, Matthew E., and Goldrath, Ananda W.
- Abstract
This corrects the article DOI: 10.1038/nature24993 [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
64. Cbx3/HP1γ deficiency confers enhanced tumor-killing capacity on CD8+ T cells.
- Author
-
Sun, Michael, Ha, Ngoc, Pham, Duc-Hung, Frederick, Megan, Sharma, Bandana, Naruse, Chie, Asano, Masahide, Pipkin, Matthew E., George, Rani E., and Thai, To-Ha
- Abstract
Cbx3/HP1γ is a histone reader whose function in the immune system is not completely understood. Here, we demonstrate that in CD8
+ T cells, Cbx3/HP1γ insufficiency leads to chromatin remodeling accompanied by enhanced Prf1, Gzmb and Ifng expression. In tumors obtained from Cbx3/HP1γ-insufficient mice or wild type mice treated with Cbx3/HP1γ-insufficient CD8+ T cells, there is an increase of CD8+ effector T cells expressing the stimulatory receptor Klrk1/NKG2D, a decrease in CD4+ CD25+ FOXP3+ regulatory T cells (Treg cells) as well as CD25+ CD4+ T cells expressing the inhibitory receptor CTLA4. Together these changes in the tumor immune environment may have mitigated tumor burden in Cbx3/HP1γ-insufficient mice or wild type mice treated with Cbx3/HP1γ-insufficient CD8+ T cells. These findings suggest that targeting Cbx3/HP1γ can represent a rational therapeutic approach to control growth of solid tumors. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
65. Erratum: Runx3 programs CD8+T cell residency in non-lymphoid tissues and tumours
- Author
-
Milner, J. Justin, Toma, Clara, Yu, Bingfei, Zhang, Kai, Omilusik, Kyla, Phan, Anthony T., Wang, Dapeng, Getzler, Adam J., Nguyen, Toan, Crotty, Shane, Wang, Wei, Pipkin, Matthew E., and Goldrath, Ananda W.
- Abstract
This corrects the article DOI: 10.1038/nature24993
- Published
- 2018
- Full Text
- View/download PDF
66. Erratum: Epigenetic landscapes reveal transcription factors that regulate CD8+T cell differentiation
- Author
-
Yu, Bingfei, Zhang, Kai, Milner, J Justin, Toma, Clara, Chen, Runqiang, Scott-Browne, James P, Pereira, Renata M, Crotty, Shane, Chang, John T, Pipkin, Matthew E, Wang, Wei, and Goldrath, Ananda W
- Published
- 2017
- Full Text
- View/download PDF
67. Kdm6b Regulates the Generation of Effector CD81 T Cells by Inducing Chromatin Accessibility in Effector-Associated Genes.
- Author
-
Tianhao Xu, Schutte, Alexander, Jimenez, Leandro, Goncalves, Andre N. A., Keller, Ashleigh, Pipkin, Matthew E., Nakaya, Helder I., Pereira, Renata M., and Martinez, Gustavo J.
- Subjects
- *
T cells , *T cell differentiation , *CHROMATIN , *EIGENFUNCTIONS , *GENES , *CYTOTOXIC T cells , *CD28 antigen - Abstract
The transcriptional and epigenetic regulation of CD8+ T cell differentiation is critical for balancing pathogen eradication and long-term immunity by effector and memory CTLs, respectively. In this study, we demonstrate that the lysine demethylase 6b (Kdm6b) is essential for the proper generation and function of effector CD81 T cells during acute infection and tumor eradication. We found that cells lacking Kdm6b (by either T cell-specific knockout mice or knockdown using short hairpin RNA strategies) show an enhanced generation of memory precursor and early effector cells upon acute viral infection in a cell-intrinsic manner. We also demonstrate that Kdm6b is indispensable for proper effector functions and tumor protection, and that memory CD8+ T cells lacking Kdm6b displayed a defective recall response. Mechanistically, we identified that Kdm6b, through induction of chromatin accessibility in key effector-associated gene loci, allows for the proper generation of effector CTLs. Our results pinpoint the essential function of Kdm6b in allowing chromatin accessibility in effector-associated genes, and identify Kdm6b as a potential target for therapeutics in diseases with dysregulated effector responses. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
68. In Vivo RNA Interference Screens Identify Regulators of Antiviral CD4+ and CD8+ T Cell Differentiation.
- Author
-
Runqiang Chen, Bélanger, Simon, Frederick, Megan A., Bin Li, Johnston, Robert J., Nengming Xiao, Yun-Cai Liu, Sharma, Sonia, Peters, Bjoern, Rao, Anjana, Crotty, Shane, and Pipkin, Matthew E.
- Subjects
- *
RNA interference , *ANTIVIRAL agents , *CD4 antigen , *CD8 antigen , *T cell differentiation , *CYCLINS - Abstract
Most genes that specify T cell differentiation during infections are unknown. Crotty, Pipkin, and colleagues develop a pooled RNA interference approach using shRNAmirs in vivo and identify the P-TEFb subunits Cyclin T1 and Cdk9 as specific regulators of antiviral T cells. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
69. Transcriptional and methylation outcomes of didehydro-cortistatin A use in HIV-1-infected CD4 + T cells.
- Author
-
Mori LP, Corley MJ, McAuley AT, Pang A, Venables T, Ndhlovu LC, Pipkin ME, and Valente ST
- Subjects
- Humans, Transcription, Genetic drug effects, Epigenesis, Genetic drug effects, Th1 Cells immunology, Th1 Cells drug effects, Th1 Cells metabolism, Isoquinolines, HIV-1 drug effects, HIV-1 physiology, CD4-Positive T-Lymphocytes metabolism, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes drug effects, HIV Infections drug therapy, HIV Infections immunology, HIV Infections virology, HIV Infections genetics, DNA Methylation drug effects, Heterocyclic Compounds, 4 or More Rings pharmacology
- Abstract
Ongoing viral transcription from the reservoir of HIV-1 infected long-lived memory CD4
+ T cells presents a barrier to cure and associates with poorer health outcomes for people living with HIV, including chronic immune activation and inflammation. We previously reported that didehydro-cortistatin A (dCA), an HIV-1 Tat inhibitor, blocks HIV-1 transcription. Here, we examine the impact of dCA on host immune CD4+ T-cell transcriptional and epigenetic states. We performed a comprehensive analysis of genome-wide transcriptomic and DNA methylation profiles upon long-term dCA treatment of primary human memory CD4+ T cells. dCA prompted specific transcriptional and DNA methylation changes in cell cycle, histone, interferon-response, and T-cell lineage transcription factor genes, through inhibition of both HIV-1 and Mediator kinases. These alterations establish a tolerogenic Treg/Th2 phenotype, reducing viral gene expression and mitigating inflammation in primary CD4+ T cells during HIV-1 infection. In addition, dCA suppresses the expression of lineage-defining transcription factors for Th17 and Th1 cells, critical HIV-1 targets, and reservoirs. dCA's benefits thus extend beyond viral transcription inhibition, modulating the immune cell landscape to limit HIV-1 acquisition and inflammatory environment linked to HIV infection., (© 2024 Mori et al.)- Published
- 2024
- Full Text
- View/download PDF
70. Spatiotemporal resolution of germinal center Tfh cell differentiation and divergence from central memory CD4 + T cell fate.
- Author
-
Zhu F, McMonigle RJ, Schroeder AR, Xia X, Figge D, Greer BD, González-Avalos E, Sialer DO, Wang YH, Chandler KM, Getzler AJ, Brown ER, Xiao C, Kutsch O, Harada Y, Pipkin ME, and Hu H
- Subjects
- Programmed Cell Death 1 Receptor genetics, Programmed Cell Death 1 Receptor metabolism, Germinal Center, Cell Differentiation, Receptors, CXCR5 genetics, Receptors, CXCR5 metabolism, T-Lymphocytes, Helper-Inducer metabolism, T Follicular Helper Cells metabolism
- Abstract
Follicular helper T (Tfh) cells are essential for germinal center (GC) B cell responses. However, it is not clear which PD-1
+ CXCR5+ Bcl6+ CD4+ T cells will differentiate into PD-1hi CXCR5hi Bcl6hi GC-Tfh cells and how GC-Tfh cell differentiation is regulated. Here, we report that the sustained Tigit expression in PD-1+ CXCR5+ CD4+ T cells marks the precursor Tfh (pre-Tfh) to GC-Tfh transition, whereas Tigit- PD-1+ CXCR5+ CD4+ T cells upregulate IL-7Rα to become CXCR5+ CD4+ T memory cells with or without CCR7. We demonstrate that pre-Tfh cells undergo substantial further differentiation at the transcriptome and chromatin accessibility levels to become GC-Tfh cells. The transcription factor c-Maf appears critical in governing the pre-Tfh to GC-Tfh transition, and we identify Plekho1 as a stage-specific downstream factor regulating the GC-Tfh competitive fitness. In summary, our work identifies an important marker and regulatory mechanism of PD-1+ CXCR5+ CD4+ T cells during their developmental choice between memory T cell fate and GC-Tfh cell differentiation., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
71. DNA architectural protein CTCF facilitates subset-specific chromatin interactions to limit the formation of memory CD8 + T cells.
- Author
-
Quon S, Yu B, Russ BE, Tsyganov K, Nguyen H, Toma C, Heeg M, Hocker JD, Milner JJ, Crotty S, Pipkin ME, Turner SJ, and Goldrath AW
- Subjects
- Humans, Binding Sites, CCCTC-Binding Factor metabolism, CD8-Positive T-Lymphocytes metabolism, DNA metabolism, Protein Binding, Chromatin, Repressor Proteins genetics, Repressor Proteins metabolism
- Abstract
Although the importance of genome organization for transcriptional regulation of cell-fate decisions and function is clear, the changes in chromatin architecture and how these impact effector and memory CD8
+ T cell differentiation remain unknown. Using Hi-C, we studied how genome configuration is integrated with CD8+ T cell differentiation during infection and investigated the role of CTCF, a key chromatin remodeler, in modulating CD8+ T cell fates through CTCF knockdown approaches and perturbation of specific CTCF-binding sites. We observed subset-specific changes in chromatin organization and CTCF binding and revealed that weak-affinity CTCF binding promotes terminal differentiation of CD8+ T cells through the regulation of transcriptional programs. Further, patients with de novo CTCF mutations had reduced expression of the terminal-effector genes in peripheral blood lymphocytes. Therefore, in addition to establishing genome architecture, CTCF regulates effector CD8+ T cell heterogeneity through altering interactions that regulate the transcription factor landscape and transcriptome., Competing Interests: Declaration of interests A.W.G. is a member of the scientific advisory board of ArsenalBio. S.J.T. is a member of the scientific advisory board for Medicago, Inc., QC, Canada. No funding from Medicago or ArsenalBio was provided for this work., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
72. Mll1 pioneers histone H3K4me3 deposition and promotes formation of CD8 + T stem cell memory.
- Author
-
Getzler AJ, Frederick MA, Milner JJ, Venables T, Diao H, Toma C, Nagaraja SD, Albao DS, Bélanger S, Tsuda SM, Kim J, Crotty S, Goldrath AW, and Pipkin ME
- Abstract
CD8
+ T cells with stem cell-like properties (TSCM ) sustain adaptive immunity to intracellular pathogens and tumors. However, the developmental origins and chromatin regulatory factors (CRFs) that establish their differentiation are unclear. Using an RNA interference screen of all CRFs we discovered the histone methylase Mll1 was required during T cell receptor (TCR) stimulation for development of a TSCM precursor state and mature memory (TMEM ) cells, but not short-lived or transitory effector cell-like states, in response to viral infections and tumors. Mll1 was essential for widespread de novo deposition of histone H3 lysine 4 trimethylation (H3K4me3) upon TCR stimulation, which accounted for 70% of all activation-induced sites in mature TMEM cells. Mll1 promoted both H3K4me3 deposition and reduced TCR-induced Pol II pausing at genes whose single-cell transcriptional dynamics explained trajectories into nascent TSCM precursor states during viral infection. Our results suggest Mll1-dependent control of Pol II elongation and H3K4me3 establishes and maintains differentiation of CD8+ TSCM cell states.- Published
- 2023
- Full Text
- View/download PDF
73. The chaperone protein p32 stabilizes HIV-1 Tat and strengthens the p-TEFb/RNAPII/TAR complex promoting HIV transcription elongation.
- Author
-
Li C, Mori LP, Lyu S, Bronson R, Getzler AJ, Pipkin ME, and Valente ST
- Subjects
- Humans, Positive Transcriptional Elongation Factor B genetics, Positive Transcriptional Elongation Factor B metabolism, tat Gene Products, Human Immunodeficiency Virus genetics, tat Gene Products, Human Immunodeficiency Virus metabolism, Transcription Factors genetics, Transcription Factors metabolism, RNA Polymerase II genetics, RNA Polymerase II metabolism, Molecular Chaperones metabolism, Transcription, Genetic, HIV Long Terminal Repeat genetics, HIV-1 physiology, HIV Infections genetics
- Abstract
HIV gene expression is modulated by the combinatorial activity of the HIV transcriptional activator, Tat, host transcription factors, and chromatin remodeling complexes. To identify host factors regulating HIV transcription, we used specific single-guide RNAs and endonuclease-deficient Cas9 to perform chromatin affinity purification of the integrated HIV promoter followed by mass spectrometry. The scaffold protein, p32, also called ASF/SF2 splicing factor-associated protein, was identified among the top enriched factors present in actively transcribing HIV promoters but absent in silenced ones. Chromatin immunoprecipitation analysis confirmed the presence of p32 on active HIV promoters and its enhanced recruitment by Tat. HIV uses Tat to efficiently recruit positive transcription elongation factor b (p-TEFb) (CDK9/CCNT1) to TAR, an RNA secondary structure that forms from the first 59 bp of HIV transcripts, to enhance RNAPII transcriptional elongation. The RNA interference of p32 significantly reduced HIV transcription in primary CD4
+ T cells and in HIV chronically infected cells, independently of either HIV splicing or p32 anti-splicing activity. Conversely, overexpression of p32 specifically increased Tat-dependent HIV transcription. p32 was found to directly interact with Tat's basic domain enhancing Tat stability and half-life. Conversely, p32 associates with Tat via N- and C-terminal domains. Likely due its scaffold properties, p32 also promoted Tat association with TAR, p-TEFb, and RNAPII enhancing Tat-dependent HIV transcription. In sum, we identified p32 as a host factor that interacts with and stabilizes Tat protein, promotes Tat-dependent transcriptional regulation, and may be explored for HIV-targeted transcriptional inhibition.- Published
- 2023
- Full Text
- View/download PDF
74. Transcriptional Control of Cell Fate Determination in Antigen-Experienced CD8 T Cells.
- Author
-
Tsuda S and Pipkin ME
- Subjects
- Cell Differentiation, Chromatin, Gene Expression Regulation, CD8-Positive T-Lymphocytes, Immunologic Memory
- Abstract
Robust immunity to intracellular infections is mediated by antigen-specific naive CD8 T cells that become activated and differentiate into phenotypically and functionally diverse subsets of effector cells, some of which terminally differentiate and others that give rise to memory cells that provide long-lived protection. This developmental system is an outstanding model with which to elucidate how regulation of chromatin structure and transcriptional control establish gene expression programs that govern cell fate determination, insights from which are likely to be useful for informing the design of immunotherapeutic approaches to engineer durable immunity to infections and tumors. A unifying framework that describes how naive CD8 T cells develop into memory cells is still outstanding. We propose a model that incorporates a common early linear path followed by divergent paths that slowly lose capacity to interconvert and discuss classical and contemporary observations that support these notions, focusing on insights from transcriptional control and chromatin regulation., (Copyright © 2022 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
75. Kdm6b Regulates the Generation of Effector CD8 + T Cells by Inducing Chromatin Accessibility in Effector-Associated Genes.
- Author
-
Xu T, Schutte A, Jimenez L, Gonçalves ANA, Keller A, Pipkin ME, Nakaya HI, Pereira RM, and Martinez GJ
- Subjects
- Animals, Cells, Cultured, Chromatin genetics, Mice, Mice, Inbred C57BL, Mice, Transgenic, CD8-Positive T-Lymphocytes immunology, Chromatin immunology, Jumonji Domain-Containing Histone Demethylases immunology
- Abstract
The transcriptional and epigenetic regulation of CD8
+ T cell differentiation is critical for balancing pathogen eradication and long-term immunity by effector and memory CTLs, respectively. In this study, we demonstrate that the lysine demethylase 6b (Kdm6b) is essential for the proper generation and function of effector CD8+ T cells during acute infection and tumor eradication. We found that cells lacking Kdm6b (by either T cell-specific knockout mice or knockdown using short hairpin RNA strategies) show an enhanced generation of memory precursor and early effector cells upon acute viral infection in a cell-intrinsic manner. We also demonstrate that Kdm6b is indispensable for proper effector functions and tumor protection, and that memory CD8+ T cells lacking Kdm6b displayed a defective recall response. Mechanistically, we identified that Kdm6b, through induction of chromatin accessibility in key effector-associated gene loci, allows for the proper generation of effector CTLs. Our results pinpoint the essential function of Kdm6b in allowing chromatin accessibility in effector-associated genes, and identify Kdm6b as a potential target for therapeutics in diseases with dysregulated effector responses., (Copyright © 2021 by The American Association of Immunologists, Inc.)- Published
- 2021
- Full Text
- View/download PDF
76. Runx3 programs CD8 + T cell residency in non-lymphoid tissues and tumours.
- Author
-
Milner JJ, Toma C, Yu B, Zhang K, Omilusik K, Phan AT, Wang D, Getzler AJ, Nguyen T, Crotty S, Wang W, Pipkin ME, and Goldrath AW
- Subjects
- Adoptive Transfer, Animals, CD8-Positive T-Lymphocytes cytology, Cell Differentiation, Cell Proliferation, Chromatin genetics, Chromatin metabolism, Core Binding Factor Alpha 3 Subunit deficiency, Core Binding Factor Alpha 3 Subunit genetics, Disease Models, Animal, Female, Gene Expression Regulation, Homeostasis, Humans, Lymphocytes, Tumor-Infiltrating metabolism, Lymphocytes, Tumor-Infiltrating pathology, Male, Melanoma genetics, Melanoma pathology, Melanoma therapy, Mice, Organ Specificity genetics, Survival Analysis, T-Lymphocyte Subsets cytology, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Core Binding Factor Alpha 3 Subunit metabolism, Immunologic Memory, Melanoma immunology, Organ Specificity immunology
- Abstract
Tissue-resident memory CD8
+ T (TRM ) cells are found at common sites of pathogen exposure, where they elicit rapid and robust protective immune responses. However, the molecular signals that control TRM cell differentiation and homeostasis are not fully understood. Here we show that mouse TRM precursor cells represent a unique CD8+ T cell subset that is distinct from the precursors of circulating memory cell populations at the levels of gene expression and chromatin accessibility. Using computational and pooled in vivo RNA interference screens, we identify the transcription factor Runx3 as a key regulator of TRM cell differentiation and homeostasis. Runx3 was required to establish TRM cell populations in diverse tissue environments, and supported the expression of crucial tissue-residency genes while suppressing genes associated with tissue egress and recirculation. Furthermore, we show that human and mouse tumour-infiltrating lymphocytes share a core tissue-residency gene-expression signature with TRM cells that is associated with Runx3 activity. In a mouse model of adoptive T cell therapy for melanoma, Runx3-deficient CD8+ tumour-infiltrating lymphocytes failed to accumulate in tumours, resulting in greater rates of tumour growth and mortality. Conversely, overexpression of Runx3 enhanced tumour-specific CD8+ T cell abundance, delayed tumour growth, and prolonged survival. In addition to establishing Runx3 as a central regulator of TRM cell differentiation, these results provide insight into the signals that promote T cell residency in non-lymphoid sites, which could be used to enhance vaccine efficacy or adoptive cell therapy treatments that target cancer.- Published
- 2017
- Full Text
- View/download PDF
77. Erratum: Epigenetic landscapes reveal transcription factors that regulate CD8 + T cell differentiation.
- Author
-
Yu B, Zhang K, Milner JJ, Toma C, Chen R, Scott-Browne JP, Pereira RM, Crotty S, Chang JT, Pipkin ME, Wang W, and Goldrath AW
- Published
- 2017
- Full Text
- View/download PDF
78. Epigenetic landscapes reveal transcription factors that regulate CD8 + T cell differentiation.
- Author
-
Yu B, Zhang K, Milner JJ, Toma C, Chen R, Scott-Browne JP, Pereira RM, Crotty S, Chang JT, Pipkin ME, Wang W, and Goldrath AW
- Subjects
- Animals, CD8-Positive T-Lymphocytes microbiology, Cell Differentiation genetics, Computational Biology, Enhancer Elements, Genetic genetics, Gene Expression Profiling, Histones metabolism, Immunologic Memory genetics, Mice, Mice, Inbred C57BL, Receptors, Glucocorticoid genetics, T-Lymphocyte Subsets microbiology, YY1 Transcription Factor genetics, CD8-Positive T-Lymphocytes physiology, Epigenesis, Genetic, Listeriosis immunology, Receptors, Glucocorticoid metabolism, T-Lymphocyte Subsets physiology, YY1 Transcription Factor metabolism
- Abstract
Dynamic changes in the expression of transcription factors (TFs) can influence the specification of distinct CD8
+ T cell fates, but the observation of equivalent expression of TFs among differentially fated precursor cells suggests additional underlying mechanisms. Here we profiled the genome-wide histone modifications, open chromatin and gene expression of naive, terminal-effector, memory-precursor and memory CD8+ T cell populations induced during the in vivo response to bacterial infection. Integration of these data suggested that the expression and binding of TFs contributed to the establishment of subset-specific enhancers during differentiation. We developed a new bioinformatics method using the PageRank algorithm to reveal key TFs that influence the generation of effector and memory populations. The TFs YY1 and Nr3c1, both constitutively expressed during CD8+ T cell differentiation, regulated the formation of terminal-effector cell fates and memory-precursor cell fates, respectively. Our data define the epigenetic landscape of differentiation intermediates and facilitate the identification of TFs with previously unappreciated roles in CD8+ T cell differentiation.- Published
- 2017
- Full Text
- View/download PDF
79. MicroRNA-directed program of cytotoxic CD8+ T-cell differentiation.
- Author
-
Trifari S, Pipkin ME, Bandukwala HS, Äijö T, Bassein J, Chen R, Martinez GJ, and Rao A
- Subjects
- Adoptive Transfer, Animals, Blotting, Western, Computational Biology, DNA Primers genetics, Granzymes metabolism, Humans, Interleukin-2 Receptor alpha Subunit metabolism, Mice, Perforin metabolism, Real-Time Polymerase Chain Reaction, Sequence Analysis, DNA, Cell Differentiation physiology, Gene Expression Regulation immunology, MicroRNAs metabolism, Ribonuclease III metabolism, Signal Transduction immunology, T-Lymphocytes, Cytotoxic physiology, Virus Diseases immunology
- Abstract
Acquisition of effector properties is a key step in the generation of cytotoxic T lymphocytes (CTLs). Here we show that inflammatory signals regulate Dicer expression in CTLs, and that deletion or depletion of Dicer in mouse or human activated CD8(+) T cells causes up-regulation of perforin, granzymes, and effector cytokines. Genome-wide analysis of microRNA (miR, miRNA) changes induced by exposure of differentiating CTLs to IL-2 and inflammatory signals identifies miR-139 and miR-150 as components of an miRNA network that controls perforin, eomesodermin, and IL-2Rα expression in differentiating CTLs and whose activity is modulated by IL-2, inflammation, and antigenic stimulation. Overall, our data show that strong IL-2R and inflammatory signals act through Dicer and miRNAs to control the cytolytic program and other aspects of effector CTL differentiation.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.