139 results on '"Roychoudhuri, Rahul"'
Search Results
102. Identification of novel regulators of developmental hematopoiesis using Endoglinregulatory elements as molecular probes
- Author
-
Nasrallah, Rabab, Fast, Eva M., Solaimani, Parham, Knezevic, Kathy, Eliades, Alexia, Patel, Rahima, Thambyrajah, Roshana, Unnikrishnan, Ashwin, Thoms, Julie, Beck, Dominik, Vink, Chris S., Smith, Aileen, Wong, Jason, Shepherd, Mairi, Kent, David, Roychoudhuri, Rahul, Paul, Fabian, Klippert, Julia, Hammes, Annette, Willnow, Thomas, Göttgens, Bertie, Dzierzak, Elaine, Zon, Leonard I., Lacaud, George, Kouskoff, Valerie, and Pimanda, John E.
- Abstract
Enhancers are the primary determinants of cell identity, and specific promoter/enhancer combinations of Endoglin (ENG) have been shown to target blood and endothelium in the embryo. Here, we generated a series of embryonic stem cell lines, each targeted with reporter constructs driven by specific promoter/enhancer combinations of ENG, to evaluate their discriminative potential and value as molecular probes of the corresponding transcriptome. The Engpromoter (P) in combination with the −8/+7/+9-kb enhancers, targeted cells in FLK1 mesoderm that were enriched for blast colony forming potential, whereas the P/−8-kb enhancer targeted TIE2+/c-KIT+/CD41− endothelial cells that were enriched for hematopoietic potential. These fractions were isolated using reporter expression and their transcriptomes profiled by RNA-seq. There was high concordance between our signatures and those from embryos with defects at corresponding stages of hematopoiesis. Of the 6 genes that were upregulated in both hemogenic mesoderm and hemogenic endothelial fractions targeted by the reporters, LRP2, a multiligand receptor, was the only gene that had not previously been associated with hematopoiesis. We show that LRP2 is indeed involved in definitive hematopoiesis and by doing so validate the use of reporter gene–coupled enhancers as probes to gain insights into transcriptional changes that facilitate cell fate transitions.
- Published
- 2016
- Full Text
- View/download PDF
103. BACH2 regulates CD8+T cell differentiation by controlling access of AP-1 factors to enhancers
- Author
-
Roychoudhuri, Rahul, Clever, David, Li, Peng, Wakabayashi, Yoshiyuki, Quinn, Kylie M, Klebanoff, Christopher A, Ji, Yun, Sukumar, Madhusudhanan, Eil, Robert L, Yu, Zhiya, Spolski, Rosanne, Palmer, Douglas C, Pan, Jenny H, Patel, Shashank J, Macallan, Derek C, Fabozzi, Giulia, Shih, Han-Yu, Kanno, Yuka, Muto, Akihiko, Zhu, Jun, Gattinoni, Luca, O'Shea, John J, Okkenhaug, Klaus, Igarashi, Kazuhiko, Leonard, Warren J, and Restifo, Nicholas P
- Abstract
T cell activation upon TCR signaling can lead to development of effector and memory cells. Roychoudhuri and colleagues show that the transcription factor BACH2 promotes memory CD8+T cell generation by blocking access to genomic regulatory sites recognized by AP-1.
- Published
- 2016
- Full Text
- View/download PDF
104. BACH2 represses effector programs to stabilize Treg-mediated immune homeostasis.
- Author
-
Roychoudhuri, Rahul, Hirahara, Kiyoshi, Mousavi, Kambiz, Clever, David, Klebanoff, Christopher A., Bonelli, Michael, Sciumè, Giuseppe, Zare, Hossein, Vahedi, Golnaz, Dema, Barbara, Yu, Zhiya, Liu, Hui, Takahashi, Hayato, Rao, Mahadev, Muranski, Pawel, Crompton, Joseph G., Punkosdy, George, Bedognetti, Davide, Wang, Ena, and Hoffmann, Victoria
- Subjects
- *
AUTOIMMUNE diseases , *HOMEOSTASIS , *ASTHMA , *CROHN'S disease , *TRANSCRIPTION factors , *T cells , *GENETIC polymorphisms , *MULTIPLE sclerosis - Abstract
Through their functional diversification, distinct lineages of CD4+ T cells can act to either drive or constrain immune-mediated pathology. Transcription factors are critical in the generation of cellular diversity, and negative regulators antagonistic to alternate fates often act in conjunction with positive regulators to stabilize lineage commitment. Genetic polymorphisms within a single locus encoding the transcription factor BACH2 are associated with numerous autoimmune and allergic diseases including asthma, Crohn's disease, coeliac disease, vitiligo, multiple sclerosis and type 1 diabetes. Although these associations point to a shared mechanism underlying susceptibility to diverse immune-mediated diseases, a function for BACH2 in the maintenance of immune homeostasis has not been established. Here, by studying mice in which the Bach2 gene is disrupted, we define BACH2 as a broad regulator of immune activation that stabilizes immunoregulatory capacity while repressing the differentiation programs of multiple effector lineages in CD4+ T cells. BACH2 was required for efficient formation of regulatory (Treg) cells and consequently for suppression of lethal inflammation in a manner that was Treg-cell-dependent. Assessment of the genome-wide function of BACH2, however, revealed that it represses genes associated with effector cell differentiation. Consequently, its absence during Treg polarization resulted in inappropriate diversion to effector lineages. In addition, BACH2 constrained full effector differentiation within TH1, TH2 and TH17 cell lineages. These findings identify BACH2 as a key regulator of CD4+ T-cell differentiation that prevents inflammatory disease by controlling the balance between tolerance and immunity. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
105. Single-cell gene-expression profiling reveals qualitatively distinct CD8 I cells elicited by different gene-based vaccines.
- Author
-
FIatz, Lukas, Roychoudhuri, Rahul, Honda, Mitsuo, Filali-Mouhim, Abdelali, Goulet, Jean-Philippe, Kettaf, Nadia, Lin, Min, Roederer, Mario, Haddad, Elias K., Sékaly, Rafick P., and Nabel, Gary J.
- Subjects
- *
DNA vaccines , *GENE expression , *IMMUNOPHENOTYPING , *IMMUNE response , *DNA microarrays , *LEISHMANIA , *INTERLEUKINS , *T cells - Abstract
CD8 T cells play a key role in mediating protective immunity against selected pathogens after vaccination. Understanding the mechanism of this protection is dependent upon definition of the heterogeneity and complexity of cellular immune responses generated by different vaccines. Here, we identify previously unrecognized subsets of CD8 T cells based upon analysis of gene-expression patterns within single cells and show that they are differentially induced by different vaccines. Three prime-boost vector combinations encoding HIV Env stimulated antigen-specific CD8 T-cell populations of similar magnitude, phenotype, and functionality. Remarkably, however, analysis of single-cell gene-expression profiles enabled discrimination of a majority of central memory (CM) and effector memory (EM) CD8 T cells elicited by the three vaccines. Subsets of T cells could be defined based on their expression of Eomes, Cxcr3, and Ccrl, or Kirki, Kirgi, and CcrS in CM and EM cells, respectively, Of CM cells elicited by DNA prime-recombinant adenoviral (rAd) boost vectors, 67% were Eomes Ccrr Cxcr3, in contrast to only 7% and 2% stimulated by rAcl5-rAd5 or rAd-LCMV, respectively, Of EM cells elicited by DNArAd, 74% were KIrkl K!rgrCcr5T compared with only 26% and 20% for rAd5-rAd5 or rAd5-LCMV. Definition by single-cell gene profiling of specific CM and EM CD8 T-cell subsets that are differentially induced by different gene-based vaccines will facilitate the design and evaluation of vaccines, as well as enable our understanding of mechanisms of protective immunity. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
106. Compensation between CSF1R+ macrophages and Foxp3+ Treg cells drives resistance to tumor immunotherapy
- Author
-
Gyori, David, Lim, Ee Lyn, Grant, Francis M, Spensberger, Dominik, Roychoudhuri, Rahul, Shuttleworth, Stephen J, Okkenhaug, Klaus, Stephens, Len R, and Hawkins, Phillip T
- Subjects
Male ,Class I Phosphatidylinositol 3-Kinases ,Immunology ,Primary Cell Culture ,T cells ,Aminopyridines ,chemical and pharmacologic phenomena ,Cancer immunotherapy ,Mice, Transgenic ,T-Lymphocytes, Regulatory ,Lymphocyte Depletion ,Gene Knockout Techniques ,Mice ,Phosphatidylinositol 3-Kinases ,stomatognathic system ,Cell Line, Tumor ,Neoplasms ,Tumor Microenvironment ,Animals ,Humans ,Diphtheria Toxin ,Pyrroles ,Phosphoinositide-3 Kinase Inhibitors ,Quinazolinones ,FOS: Clinical medicine ,Macrophages ,hemic and immune systems ,Forkhead Transcription Factors ,3. Good health ,Disease Models, Animal ,Oncology ,Drug Resistance, Neoplasm ,Purines ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,Female - Abstract
Redundancy and compensation provide robustness to biological systems but may contribute to therapy resistance. Both tumor-associated macrophages (TAMs) and Foxp3+ regulatory T (Treg) cells promote tumor progression by limiting antitumor immunity. Here we show that genetic ablation of CSF1 in colorectal cancer cells reduces the influx of immunosuppressive CSF1R+ TAMs within tumors. This reduction in CSF1-dependent TAMs resulted in increased CD8+ T cell attack on tumors, but its effect on tumor growth was limited by a compensatory increase in Foxp3+ Treg cells. Similarly, disruption of Treg cell activity through their experimental ablation produced moderate effects on tumor growth and was associated with elevated numbers of CSF1R+ TAMs. Importantly, codepletion of CSF1R+ TAMs and Foxp3+ Treg cells resulted in an increased influx of CD8+ T cells, augmentation of their function, and a synergistic reduction in tumor growth. Further, inhibition of Treg cell activity either through systemic pharmacological blockade of PI3Kδ, or its genetic inactivation within Foxp3+ Treg cells, sensitized previously unresponsive solid tumors to CSF1R+ TAM depletion and enhanced the effect of CSF1R blockade. These findings identify CSF1R+ TAMs and PI3Kδ-driven Foxp3+ Treg cells as the dominant compensatory cellular components of the immunosuppressive tumor microenvironment, with implications for the design of combinatorial immunotherapies.
107. Regulation of regulatory T cells in cancer
- Author
-
Stockis, Julie, Roychoudhuri, Rahul, and Halim, Timotheus YF
- Subjects
tumour immunology ,chemical and pharmacologic phenomena ,T-Lymphocytes, Regulatory ,regulatory T cells ,3. Good health ,Lymphocytes, Tumor-Infiltrating ,Phenotype ,cytokines/cytokine receptors ,Neoplasms ,chemokine/chemokine receptors ,Tumor Microenvironment ,cancer ,Animals ,Humans ,Tumor Escape ,Inflammation Mediators ,Signal Transduction - Abstract
The inflammatory response to transformed cells forms the cornerstone of natural or therapeutically induced protective immunity to cancer. Regulatory T (Treg) cells are known for their critical role in suppressing inflammation, and therefore can antagonize effective anti-cancer immune responses. As such, Treg cells can play detrimental roles in tumour progression and in the response to both conventional and immune-based cancer therapies. Recent advances in our understanding of Treg cells reveal complex niche-specific regulatory programmes and functions, which are likely to extrapolate to cancer. The regulation of Treg cells is reliant on upstream cues from haematopoietic and non-immune cells, which dictates their genetic, epigenetic and downstream functional programmes. In this review we will discuss how Treg cells are themselves regulated in normal and transformed tissues, and the implications of this cross talk on tumour growth.
108. Ionic immune suppression within the tumour microenvironment limits T cell effector function
- Author
-
Eil, Robert, Vodnala, Suman K, Clever, David, Klebanoff, Christopher A, Sukumar, Madhusudhanan, Pan, Jenny H, Palmer, Douglas C, Gros, Alena, Yamamoto, Tori N, Patel, Shashank J, Guittard, Geoffrey C, Yu, Zhiya, Carbonaro, Valentina, Okkenhaug, Klaus, Schrump, David S, Linehan, W Marston, Roychoudhuri, Rahul, and Restifo, Nicholas P
- Subjects
Male ,Kv1.3 Potassium Channel ,T-Lymphocytes ,TOR Serine-Threonine Kinases ,Receptors, Antigen, T-Cell ,Cations, Monovalent ,Survival Analysis ,3. Good health ,Membrane Potentials ,Mice ,Necrosis ,Immune Tolerance ,Potassium ,Tumor Microenvironment ,Animals ,Humans ,Tumor Escape ,Immunotherapy ,Melanoma ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Tumours progress despite being infiltrated by tumour-specific effector T cells. Tumours contain areas of cellular necrosis, which are associated with poor survival in a variety of cancers. Here, we show that necrosis releases intracellular potassium ions into the extracellular fluid of mouse and human tumours, causing profound suppression of T cell effector function. Elevation of the extracellular potassium concentration ([K+]e) impairs T cell receptor (TCR)-driven Akt-mTOR phosphorylation and effector programmes. Potassium-mediated suppression of Akt-mTOR signalling and T cell function is dependent upon the activity of the serine/threonine phosphatase PP2A. Although the suppressive effect mediated by elevated [K+]e is independent of changes in plasma membrane potential (Vm), it requires an increase in intracellular potassium ([K+]i). Accordingly, augmenting potassium efflux in tumour-specific T cells by overexpressing the potassium channel Kv1.3 lowers [K+]i and improves effector functions in vitro and in vivo and enhances tumour clearance and survival in melanoma-bearing mice. These results uncover an ionic checkpoint that blocks T cell function in tumours and identify potential new strategies for cancer immunotherapy.
109. The transcriptional programme of natural killer (NK) cell functional maturation and maintenance
- Author
-
Imianowski, Charlotte and Roychoudhuri, Rahul
- Subjects
Natural Killer cells ,Innate lymphocytes ,Anti-tumour immunity ,Immune surveillance ,Transcriptional regulation ,Transcription factor ,BACH2 - Abstract
Natural killer (NK) cells are critical to immune surveillance against infections and cancer. Their role in immune surveillance requires that NK cells are present within tissues in a quiescent state. The functional maturation of NK cells is a tightly regulated process which is controlled by transcription factors (TFs), and multiple positive regulators of this process have been defined. However, mechanisms by which NK cells remain quiescent in tissues are incompletely elucidated. The transcriptional repressor BACH2 plays a critical role within the adaptive immune system but its function within innate lymphocytes has been unclear. The studies presented here show that BACH2 acts as an intrinsic negative regulator of NK cell maturation and function. BACH2 is expressed within developing and mature NK cells and promotes the maintenance of immature NK cells by restricting their maturation in the presence of tonic IL-15 signalling. Loss of BACH2 within NK cells results in accumulation of activated NK cells with unrestrained cytotoxic function and increased immune surveillance to pulmonary cancer metastasis. These findings establish a critical function of BACH2 as a negative regulator of innate cytotoxic function and tumour immune surveillance by NK cells.
- Published
- 2022
- Full Text
- View/download PDF
110. Control of chromatin accessibility during CD4+ T cell development and activation
- Author
-
Sadiyah, Mohammed Firas and Roychoudhuri, Rahul
- Subjects
T-cell ,Immunology ,Genomics ,Epigenetics - Abstract
T cells coordinate immune functions by differentiating into highly specialised cellular lineages that either promote or suppress immune reactions. Whereas effector T (Teff) cells drive immune activation and promote clearance of infections and cancer, regulatory T (Treg) cells suppress their function to prevent excessive immune reactions that would otherwise result in autoimmune and allergic disease. During cell fate decisions, access to lineage-specific regulatory elements in the genome, which is tightly controlled by chromatin structure, is required to acquire a specific gene expression profile and a new cell identity. The dynamic changes of the chromatin landscape during Treg differentiation are largely unknown. To study this, I measured chromatin accessibility during the activation of naïve CD4 + T cells in the presence of TGF β (iTreg) or no cytokine (Th0) at different time points and integrated them with known gene expression and histone modification datasets to further characterise these changes. This shows that the chromatin landscape in iTreg cells undergoes dynamic remodelling in response to TCR stimulation and cytokine signalling with convergent effects of the two signals at many regulated loci. BACH2 is a transcriptional repressor whose expression is predominantly restricted to lymphocytes. Whether the suppression role of BACH2 is mainly through steric competition with the AP-1 family or involves other mechanisms is currently unclear. To test this, I measured gene-wide accessibility, DNA binding, and gene expression in Bach2-deficient and Bach2-sufficient Treg cells. While BACH2 is a critical regulator of T cell differentiation, its effect on accessible chromatin was minimal supporting a model whereby it functions principally as a steric repressor of AP-1-driven gene expression. Prior to Treg lineage commitment, BACH2 expression is required for induction of Foxp3, but its role after Treg lineage commitment has been unclear. To test this, I examined gene expression and genome-wide chromatin accessibility in natural Treg (nTreg) cells. This shows that BACH2 is repurposed following Treg lineage commitment, and represses the induction of activated Treg genes to promote the quiescence of resting Treg cells.
- Published
- 2021
111. Mechanisms of CD4+ regulatory T cell heterogeneity in health and disease
- Author
-
Grant, Francis Matthew and Roychoudhuri, Rahul
- Subjects
616.07 ,T cell ,immunosuppression ,autoimmune disease ,cancer ,immunology ,regulatory T cell ,transcription factor - Abstract
Regulatory T (Treg) cells are central to the maintenance of immune homeostasis and their dysfunction underlies the pathology of numerous diseases. Treg cell populations are phenotypically heterogeneous, comprising functionally quiescent resting Treg (rTreg) cells, which upon antigen stimulation, differentiate into functionally activated Treg (aTreg) cells. The purpose of rTreg cell populations and how their naïve-like phenotype is maintained despite chronic exposure to cognate self- and foreign antigens remains to be understood. The transcription factor BACH2 is critical for early Treg cell lineage specification, however, its function following Treg lineage-commitment is unresolved. The studies detailed herein demonstrate that Bach2 is highly expressed during Treg cell development in the thymus. High levels of Bach2 are maintained in post-thymic, lineage-committed rTreg cells but is downregulated in aTreg cells, and upon inflammation. Functionally, BACH2 acts to restrain T cell receptor (TCR)-driven activation in rTreg cells and constrain their differentiation into aTreg cells. Cell-autonomous expression of BACH2 is required following Treg cell lineage-commitment for functional quiescence and long-term maintenance of Treg cell populations. This is necessary for the restraint of excessive memory differentiation and IFN-γ production by CD8+ T cells. Therefore, in lineage-committed Treg cells, BACH2-mediated restraint of aTreg cell differentiation is required for the maintenance of immune homeostasis. These findings deepen our understanding of Treg cell biology and extend our knowledge of the function of the transcription factor BACH2 in lymphocytes.
- Published
- 2020
- Full Text
- View/download PDF
112. TCR-induced FOXP3 expression by CD8+ T cells impairs their anti-tumor activity.
- Author
-
Lozano, Teresa, Conde, Enrique, Martín-Otal, Celia, Navarro, Flor, Lasarte-Cia, Aritz, Nasrallah, Rabab, Alignani, Diego, Gorraiz, Marta, Sarobe, Pablo, Romero, Juan P., Vilas, Amaia, Roychoudhuri, Rahul, Hervás-Stubbs, Sandra, Casares, Noelia, and Lasarte, Juan José
- Subjects
- *
T cells , *ANTINEOPLASTIC agents , *T cell receptors , *CELL analysis , *TRANSCRIPTION factors , *PROTEIN metabolism , *RESEARCH , *IMMUNIZATION , *ANIMAL experimentation , *RESEARCH methodology , *CELL receptors , *MEDICAL cooperation , *EVALUATION research , *COMPARATIVE studies , *MICE - Abstract
Adoptive cell transfer therapy using CD8+ T lymphocytes showed promising results eradicating metastatic malignancies. However, several regulatory mechanisms limit its efficacy. We studied the role of the expression of the transcription factor FOXP3 on CD8+ T cell function and anti-tumor immunity. Here we show that suboptimal T cell receptor stimulation of CD8+ T cells upregulates FOXP3 in vitro. Similarly, CD8 T cells transferred into tumor-bearing mice upregulate FOXP3 in vivo. Cell-intrinsic loss of FOXP3 by CD8+ T cells resulted in improved functionality after TCR stimulation and better antitumor responses in vivo. Inhibition of the FOXP3/NFAT interaction likewise improved CD8+ T cell functionality. Transcriptomic analysis of cells after TCR stimulation revealed an enrichment of genes implicated in the response to IFN-γ, IFN-α, inflammatory response, IL-6/JAK/STAT, G2M checkpoint and IL-2/STAT signaling in FOXP3-deficient CD8+ T cells with respect to FOXP3-wt CD8+ T cells. Our results suggest that transient expression of FOXP3 by CD8+ T cells in the tumor microenvironment restrains their anti-tumor activity, with clear implications for improving T cell responses during immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
113. Bach2 Promotes B Cell Receptor-Induced Proliferation of B Lymphocytes and Represses Cyclin-Dependent Kinase Inhibitors.
- Author
-
Mizuho Morooka, Sax, Nicolas, Brydun, Andrey, Mitsuyo Matsumoto, Yuichi Miura, Ari Itoh-Nakadai, Akihiko Muto, Kazuhiko Igarashi, Susumu Satomi, Roychoudhuri, Rahul, Ryo Funayama, and Keiko Nakayama
- Subjects
- *
B cells , *LYMPHOCYTES , *CYCLINS , *KINASE inhibitors , *GENES - Abstract
BTB and CNC homology 2 (Bach2) is a transcriptional repressor that is required for the formation of the germinal center (GC) and reactions, including class switch recombination and somatic hypermutation of Ig genes in B cells, within the GC. Although BCR-induced proliferation is essential for GC reactions, the function of Bach2 in regulating B cell proliferation has not been elucidated. In this study, we demonstrate that Bach2 is required to sustain high levels of B cell proliferation in response to BCR signaling. Following BCR engagement in vitro, B cells from Bach2-deficient (Bach2-/-) mice showed lower incorporation of BrdU and reduced cell cycle progression compared with wild-type cells. Bach2-/- B cells also underwent increased apoptosis, as evidenced by an elevated frequency of sub-G1 cells and early apoptotic cells. Transcriptome analysis of BCR-engaged B cells from Bach2-/- mice revealed reduced expression of the antiapoptotic gene Bcl2l1 encoding Bcl-xL and elevated expression of cyclin-dependent kinase inhibitor (CKI) family genes, including Cdkn1a, Cdkn2a, and Cdkn2b. Reconstitution of Bcl-xL expression partially rescued the proliferation defect of Bach2-/- B cells. Chromatin immunoprecipitation experiments showed that Bach2 bound to the CKI family genes, indicating that these genes are direct repression targets of Bach2. These findings identify Bach2 as a requisite factor for sustaining high levels of BCR-induced proliferation, survival, and cell cycle progression, and it promotes expression of Bcl-xL and repression of CKI genes. BCR-induced proliferation defects may contribute to the impaired GC formation observed in Bach2-/- mice. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
114. CCR8 marks highly suppressive Treg cells within tumours but is dispensable for their accumulation and suppressive function
- Author
-
Sarah K. Whiteside, Jie Yang, Frank Tacke, Alberto G. Conti, Charlotte J. Imianowski, Robert L. Eil, James Dooley, Adrian Liston, Rabab Nasrallah, Rahul Roychoudhuri, Francis M. Grant, Firas Sadiyah, Paula Kuo, Oliver T. Burton, Klaus Okkenhaug, David Gyori, Sergio A. Lira, Whiteside, Sarah K [0000-0003-4354-4713], Okkenhaug, Klaus [0000-0002-9432-4051], Roychoudhuri, Rahul [0000-0002-5392-1853], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Chemokine ,Skin Neoplasms ,medicine.medical_treatment ,Immunology ,Melanoma, Experimental ,Context (language use) ,chemical and pharmacologic phenomena ,CCR8 ,Biology ,Adenocarcinoma ,CD8+ T cells ,T-Lymphocytes, Regulatory ,Receptors, CCR8 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Lymphocytes, Tumor-Infiltrating ,medicine ,Immune Tolerance ,Immunology and Allergy ,Cytotoxic T cell ,cancer ,Animals ,Humans ,Melanoma ,Mice, Knockout ,FOXP3 ,Immunosuppression ,hemic and immune systems ,Forkhead Transcription Factors ,Immunotherapy ,Original Articles ,CD4+ T cells ,Treg ,Mice, Inbred C57BL ,030104 developmental biology ,Foxp3 ,Cancer research ,biology.protein ,Original Article ,immunotherapy ,Antibody ,Colorectal Neoplasms ,030215 immunology - Abstract
CD4+ regulatory T (Treg) cells, dependent upon the transcription factor Foxp3, contribute to tumour immunosuppression but are also required for immune homeostasis. There is interest in developing therapies that selectively target the immunosuppressive function of Treg cells within tumours without disrupting their systemic anti‐inflammatory function. High levels of expression of chemokine (C‐C motif) receptor 8 (CCR8) discriminate Treg cells within tumours from those found in systemic lymphoid tissues. It has recently been proposed that disruption of CCR8 function using blocking anti‐CCR8 antibodies results in reduced accumulation of Treg cells within tumours and disruption of their immunosuppressive function. Here, using Ccr8 −/− mice, we show that CCR8 function is not required for Treg cell accumulation or immunosuppression in the context of syngeneic MC38 colorectal adenocarcinoma and B16 melanoma tumours. We observed high levels of CCR8 expression on tumour‐infiltrating Treg cells which were abolished in Ccr8 −/− mice. High levels of CCR8 marked cells with high levels of suppressive function. However, whereas systemic ablation of Treg cells resulted in strikingly diminished tumour burden, growth of subcutaneously implanted tumours was unaffected by systemic CCR8 loss. Consistently, we observed minimal impact of systemic CCR8 ablation on the frequency, phenotype and function of tumour‐infiltrating Treg cells and conventional T (Tconv) function. These findings suggest that CCR8 is not required for Treg cell accumulation and immunosuppressive function within tumours and that depletion of CCR8+ Treg cells rather than blockade of CCR8 function is a more promising avenue for selective immunotherapy., There is interest in developing therapies that selectively target the immunosuppressive function of Treg cells within tumours without disrupting their systemic anti‐inflammatory function. It has recently been proposed that disruption of CCR8 function using blocking anti‐CCR8 antibodies results in reduced accumulation of Treg cells within tumours and disruption of their immunosuppressive function.We show that CCR8 marks highly suppressive Treg cells within tumours but is not required for Treg cell accumulation and immunosuppressive function within tumours, suggesting that depletion of CCR8+ Treg cells rather than blockade of CCR8 function is a more promising avenue for selective immunotherapy.
- Published
- 2021
115. A cell-based bioluminescence assay reveals dose-dependent and contextual repression of AP-1-driven gene expression by BACH2
- Author
-
Charlotte J. Imianowski, Rahul Roychoudhuri, Jie Yang, Christopher Bot, Paula Kuo, Teresa Lozano, Simon Walker, Klaus Okkenhaug, Hanneke Okkenhaug, Panagiota Vardaka, Jonathan Ellery, Stuart Farrow, Sarah K. Whiteside, Apollo - University of Cambridge Repository, Walker, Simon [0000-0001-9185-4922], Yang, Jie [0000-0001-9157-0532], Okkenhaug, Klaus [0000-0002-9432-4051], Kuo, Paula [0000-0002-2476-0018], and Roychoudhuri, Rahul [0000-0002-5392-1853]
- Subjects
631/67 ,Cell biology ,Molecular biology ,T cell ,Immunology ,lcsh:Medicine ,Phenylenediamines ,Jurkat Cells ,Mice ,Gene expression ,medicine ,Animals ,Humans ,Luciferase ,Luciferases ,lcsh:Science ,Enhancer ,Transcription factor ,Cancer ,Acrylamides ,Reporter gene ,Multidisciplinary ,631/1647 ,Dose-Response Relationship, Drug ,Effector ,Chemistry ,631/250 ,Biological techniques ,lcsh:R ,article ,Cell Differentiation ,Tetracycline ,HDAC3 ,Transcription Factor AP-1 ,Basic-Leucine Zipper Transcription Factors ,medicine.anatomical_structure ,Gene Expression Regulation ,Luminescent Measurements ,Tetradecanoylphorbol Acetate ,lcsh:Q ,631/80 ,631/337 - Abstract
Whereas effector CD4+ and CD8+ T cells promote immune activation and can drive clearance of infections and cancer, CD4+ regulatory T (Treg) cells suppress their function, contributing to both immune homeostasis and cancer immunosuppression. The transcription factor BACH2 functions as a pervasive regulator of T cell differentiation, promoting development of CD4+ Treg cells and suppressing the effector functions of multiple effector T cell (Teff) lineages. Here, we report the development of a stable cell-based bioluminescence assay of the transcription factor activity of BACH2. Tetracycline-inducible BACH2 expression resulted in suppression of phorbol 12-myristate 13-acetate (PMA)/ionomycin-driven activation of a luciferase reporter containing BACH2/AP-1 target sequences from the mouse Ifng + 18k enhancer. BACH2 expression repressed the luciferase signal in a dose-dependent manner but this activity was abolished at high levels of AP-1 signalling, suggesting contextual regulation of AP-1 driven gene expression by BACH2. Finally, using the reporter assay developed, we find that the histone deacetylase 3 (HDAC3)-selective inhibitor, RGFP966, inhibits BACH2-mediated repression of signal-driven luciferase expression. In addition to enabling mechanistic studies, this cell-based reporter may enable identification of small molecule agonists or antagonists of BACH2 function for drug development.
- Published
- 2020
116. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function.
- Author
-
Sukumar, Madhusudhanan, Jie Liu, Yun Ji, Subramanian, Murugan, Crompton, Joseph G., Zhiya Yu, Roychoudhuri, Rahul, Palmer, Douglas C., Muranski, Pawel, Karoly, Edward D., Mohney, Robert P., Klebanoff, Christopher A., Lal, Ashish, Finkel, Toren, Restifo, Nicholas P., and Gattinoni, Luca
- Subjects
- *
T cells , *OXIDATION , *PHYSIOLOGICAL effects of fatty acids , *ANTIGENS , *GLUCOSE metabolism - Abstract
Naive CD8+ T cells rely upon oxidation of fatty acids as a primary source of energy. After antigen encounter, T cells shift to a glycolytic metabolism to sustain effector function. It is unclear, however, whether changes in glucose metabolism ultimately influence the ability of activated T cells to become long-lived memory cells. We used a fluorescent glucose analog, 2-NBDG, to quantify glucose uptake in activated CD8+ T cells. We found that cells exhibiting limited glucose incorporation had a molecular profile characteristic of memory precursor cells and an increased capacity to enter the memory pool compared with cells taking up high amounts of glucose. Accordingly, enforcing glycolytic metabolism by overexpressing the glycolytic enzyme phosphoglycerate mutase-1 severely impaired the ability of CD8+ T cells to form long-term memory. Conversely, activation of CD8+ T cells in the presence of an inhibitor of glycolysis, 2-deoxyglucose, enhanced the generation of memory cells and antitumor functionality. Our data indicate that augmenting glycolytic flux drives CD8+ T cells toward a terminally differentiated state, while its inhibition preserves the formation of long-lived memory CD8+ T cells. These results have important implications for improving the efficacy of T cell-based therapies against chronic infectious diseases and cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
117. Regulation of regulatory T cells in cancer
- Author
-
Julie Stockis, Timotheus Y.F. Halim, Rahul Roychoudhuri, Stockis, Julie [0000-0003-4911-6891], Roychoudhuri, Rahul [0000-0002-5392-1853], Halim, Timotheus YF [0000-0001-9773-0023], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Protective immunity ,Inflammatory response ,Immunology ,Inflammation ,chemical and pharmacologic phenomena ,Review Article ,Biology ,Treg cell ,T-Lymphocytes, Regulatory ,regulatory T cells ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Lymphocytes, Tumor-Infiltrating ,Review Series: Tregs in Cancer: Where are we now? ,Neoplasms ,medicine ,chemokine/chemokine receptors ,Tumor Microenvironment ,Immunology and Allergy ,cancer ,Animals ,Humans ,Epigenetics ,Review Articles ,tumour immunology ,Cancer ,medicine.disease ,3. Good health ,Haematopoiesis ,030104 developmental biology ,Phenotype ,cytokines/cytokine receptors ,Series Editors: Awen Gallimore, Sergio Quezada & Rahul Roychoudhuri ,Cancer research ,Tumor Escape ,medicine.symptom ,Inflammation Mediators ,030215 immunology ,Signal Transduction - Abstract
Summary The inflammatory response to transformed cells forms the cornerstone of natural or therapeutically induced protective immunity to cancer. Regulatory T (Treg) cells are known for their critical role in suppressing inflammation, and therefore can antagonize effective anti‐cancer immune responses. As such, Treg cells can play detrimental roles in tumour progression and in the response to both conventional and immune‐based cancer therapies. Recent advances in our understanding of Treg cells reveal complex niche‐specific regulatory programmes and functions, which are likely to extrapolate to cancer. The regulation of Treg cells is reliant on upstream cues from haematopoietic and non‐immune cells, which dictates their genetic, epigenetic and downstream functional programmes. In this review we will discuss how Treg cells are themselves regulated in normal and transformed tissues, and the implications of this cross talk on tumour growth.
- Published
- 2019
118. BACH2 regulates CD8(+) T cell differentiation by controlling access of AP-1 factors to enhancers
- Author
-
Klaus Okkenhaug, Yuka Kanno, Luca Gattinoni, Han-Yu Shih, Peng Li, Giulia Fabozzi, Kylie M. Quinn, Yun Ji, Derek C. Macallan, Jun Zhu, Yoshiyuki Wakabayashi, Shashank J. Patel, Warren J. Leonard, Robert L. Eil, Akihiko Muto, Zhiya Yu, Christopher A. Klebanoff, Kazuhiko Igarashi, Douglas C. Palmer, Rosanne Spolski, Rahul Roychoudhuri, Jenny H. Pan, David Clever, Madhusudhanan Sukumar, Nicholas P. Restifo, John J. O'Shea, Roychoudhuri, Rahul [0000-0002-5392-1853], Okkenhaug, Klaus [0000-0002-9432-4051], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Cellular differentiation ,Immunology ,Oncogene Protein p65(gag-jun) ,Vaccinia virus ,Biology ,Adaptive Immunity ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Article ,03 medical and health sciences ,Mice ,Vaccinia ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Enhancer ,Cells, Cultured ,Mice, Knockout ,Effector ,Activator (genetics) ,T-cell receptor ,Cell Differentiation ,Acquired immune system ,Mice, Inbred C57BL ,Transcription Factor AP-1 ,030104 developmental biology ,Basic-Leucine Zipper Transcription Factors ,Enhancer Elements, Genetic ,Gene Expression Regulation ,Cancer research ,Signal transduction ,Immunologic Memory ,Signal Transduction - Abstract
T cell antigen receptor (TCR) signaling drives distinct responses depending upon the differentiation state and context of CD8+ T cells. We hypothesized that access of signal-dependent transcription factors (TFs) to enhancers is dynamically regulated to shape transcriptional responses to TCR signaling. We found that the TF BACH2 restrains terminal differentiation to enable generation of long-lived memory cells and protective immunity following viral infection. BACH2 was recruited to enhancers where it limited expression of TCR-driven genes by attenuating the availability of activator protein 1 (AP-1) sites to Jun family signal-dependent TFs. In naïve cells, this prevented TCR-driven induction of genes associated with terminal differentiation. Upon effector differentiation, reduced expression of BACH2 and its phosphorylation enabled unrestrained induction of TCR-driven effector programs.
- Published
- 2018
- Full Text
- View/download PDF
119. Compensation between CSF1R+ macrophages and Foxp3+ Treg cells drives resistance to tumor immunotherapy
- Author
-
Stephen J Shuttleworth, Ee Lyn Lim, David Gyori, Dominik Spensberger, Rahul Roychoudhuri, L. Stephens, Phillip T. Hawkins, Klaus Okkenhaug, Francis M. Grant, Roychoudhuri, Rahul [0000-0002-5392-1853], Okkenhaug, Klaus [0000-0002-9432-4051], Hawkins, Phillip Thomas [0000-0002-6979-0464], and Apollo - University of Cambridge Repository
- Subjects
Male ,0301 basic medicine ,medicine.medical_treatment ,Aminopyridines ,Cancer immunotherapy ,T-Lymphocytes, Regulatory ,Gene Knockout Techniques ,Mice ,Phosphatidylinositol 3-Kinases ,Neoplasms ,Tumor Microenvironment ,Diphtheria Toxin ,Phosphoinositide-3 Kinase Inhibitors ,Chemistry ,FOXP3 ,Forkhead Transcription Factors ,hemic and immune systems ,General Medicine ,3. Good health ,medicine.anatomical_structure ,Oncology ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,Female ,Research Article ,Class I Phosphatidylinositol 3-Kinases ,T cell ,Immunology ,Primary Cell Culture ,T cells ,Mice, Transgenic ,chemical and pharmacologic phenomena ,Lymphocyte Depletion ,03 medical and health sciences ,stomatognathic system ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Pyrroles ,Quinazolinones ,Tumor microenvironment ,Macrophages ,Immunotherapy ,Disease Models, Animal ,030104 developmental biology ,Drug Resistance, Neoplasm ,Purines ,Tumor progression ,Cell culture ,Cancer research ,CD8 - Abstract
Redundancy and compensation provide robustness to biological systems but may contribute to therapy resistance. Both tumor-associated macrophages (TAMs) and Foxp3+ regulatory T (Treg) cells promote tumor progression by limiting antitumor immunity. Here we show that genetic ablation of CSF1 in colorectal cancer cells reduces the influx of immunosuppressive CSF1R+ TAMs within tumors. This reduction in CSF1-dependent TAMs resulted in increased CD8+ T cell attack on tumors, but its effect on tumor growth was limited by a compensatory increase in Foxp3+ Treg cells. Similarly, disruption of Treg cell activity through their experimental ablation produced moderate effects on tumor growth and was associated with elevated numbers of CSF1R+ TAMs. Importantly, codepletion of CSF1R+ TAMs and Foxp3+ Treg cells resulted in an increased influx of CD8+ T cells, augmentation of their function, and a synergistic reduction in tumor growth. Further, inhibition of Treg cell activity either through systemic pharmacological blockade of PI3Kδ, or its genetic inactivation within Foxp3+ Treg cells, sensitized previously unresponsive solid tumors to CSF1R+ TAM depletion and enhanced the effect of CSF1R blockade. These findings identify CSF1R+ TAMs and PI3Kδ-driven Foxp3+ Treg cells as the dominant compensatory cellular components of the immunosuppressive tumor microenvironment, with implications for the design of combinatorial immunotherapies.
- Published
- 2018
120. BACH2 immunodeficiency illustrates an association between super-enhancers and haploinsufficiency
- Author
-
Mohammed F Sadiyah, Ira Palmer, Anwar A. Sayed, Ahmad Khoder, Joshua Kaufman, Warren Strober, Alejandro V. Villarino, Paul T. Wingfield, Michael Mueller, Majid Kazemian, Joshua McElwee, Ahmed N. Hegazy, Behdad Afzali, Fred P. Davis, Julia Keith, Jason D. Hughes, Hong-Wei Sun, Charlotte O'Brien, Holm H. Uhlig, Michelle A. Linterman, Yu Zhang, Arian Laurence, Nichola Cooper, John J. O'Shea, Ine Vanderleyden, Dalia Kasperaviciute, Timothy J. Aitman, Rahul Roychoudhuri, Olena Kamenyeva, Juha Grönholm, Kim Montgomery-Recht, Ivan J. Fuss, Nicholas P. Restifo, Norman R. Watts, Peter Kelleher, Jana Vandrovcova, Michael J. Lenardo, Westminster Medical School Research Trust, Imperial College Healthcare NHS Trust- BRC Funding, Leuka, Zhang, Yu [0000-0002-6904-0372], Villarino, Alejandro V [0000-0001-8068-2176], Roychoudhuri, Rahul [0000-0002-5392-1853], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Male ,Pancytopenia ,AUTOIMMUNITY ,Genome-wide association study ,CELL DIFFERENTIATION ,Haploinsufficiency ,Adrenal Cortex Hormones ,Recurrence ,Plasma cell differentiation ,Immunology and Allergy ,Respiratory Tract Infections ,Immunodeficiency ,Genetics ,Lymphocyte differentiation ,Syndrome ,Middle Aged ,Colitis ,3. Good health ,Pedigree ,TRANSCRIPTION FACTORS ,Basic-Leucine Zipper Transcription Factors ,PLASMA-CELL-DIFFERENTIATION ,1107 Immunology ,B-CELLS ,Female ,FUNCTIONAL PREDICTIONS ,Life Sciences & Biomedicine ,Adult ,Heterozygote ,SUSCEPTIBILITY LOCI ,SEQUENCING DATA ,Fever ,DATABASE ,Immunology ,COMMON VARIABLE IMMUNODEFICIENCY ,Polymorphism, Single Nucleotide ,Article ,CLASSIFICATION ,Autoimmune Diseases ,03 medical and health sciences ,Young Adult ,Lymphopenia ,Journal Article ,medicine ,Humans ,GENOME-WIDE ASSOCIATION ,Gene ,METAANALYSIS ,Science & Technology ,IDENTIFICATION ,business.industry ,Common variable immunodeficiency ,Immunologic Deficiency Syndromes ,medicine.disease ,RISK LOCI ,030104 developmental biology ,Mutation ,Splenomegaly ,Primary immunodeficiency ,T-CELLS ,business ,Tomography, X-Ray Computed - Abstract
The transcriptional programs that guide lymphocyte differentiation depend on the precise expression and timing of transcription factors (TFs). The TF BACH2 is essential for T and B lymphocytes and is associated with an archetypal super-enhancer (SE). Single-nucleotide variants in the BACH2 locus are associated with several autoimmune diseases, but BACH2 mutations that cause Mendelian monogenic primary immunodeficiency have not previously been identified. Here we describe a syndrome of BACH2-related immunodeficiency and autoimmunity (BRIDA) that results from BACH2 haploinsufficiency. Affected subjects had lymphocyte-maturation defects that caused immunoglobulin deficiency and intestinal inflammation. The mutations disrupted protein stability by interfering with homodimerization or by causing aggregation. We observed analogous lymphocyte defects in Bach2-heterozygous mice. More generally, we observed that genes that cause monogenic haploinsufficient diseases were substantially enriched for TFs and SE architecture. These findings reveal a previously unrecognized feature of SE architecture in Mendelian diseases of immunity: heterozygous mutations in SE-regulated genes identified by whole-exome/genome sequencing may have greater significance than previously recognized.
- Published
- 2017
121. Identification of novel regulators of developmental hematopoiesis using Endoglin regulatory elements as molecular probes
- Author
-
Mairi Shepherd, Julie A. I. Thoms, Kathy Knezevic, Ashwin Unnikrishnan, Rahul Roychoudhuri, Berthold Göttgens, Thomas E. Willnow, Roshana Thambyrajah, Aileen M. Smith, George Lacaud, Julia Klippert, Parham Solaimani, Alexia Eliades, Rabab Nasrallah, Dominik Beck, Elaine Dzierzak, John E. Pimanda, Leonard I. Zon, Jason W. H. Wong, Annette Hammes, Eva M. Fast, Fabian Paul, Chris S. Vink, Valerie Kouskoff, David G. Kent, Rahima Patel, Cell biology, Shepherd, Mairi [0000-0002-4328-9882], Kent, David [0000-0001-7871-8811], Roychoudhuri, Rahul [0000-0002-5392-1853], Gottgens, Berthold [0000-0001-6302-5705], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Mesoderm ,Immunology ,Biology ,Cell fate determination ,Biochemistry ,Cell Line ,Transcriptome ,03 medical and health sciences ,Mice ,medicine ,Animals ,Enhancer ,Gene ,Endoglin ,Endothelial Cells ,Mouse Embryonic Stem Cells ,Cell Biology ,Hematology ,Embryo, Mammalian ,Embryonic stem cell ,Molecular biology ,Hematopoiesis ,Low Density Lipoprotein Receptor-Related Protein-2 ,030104 developmental biology ,medicine.anatomical_structure ,Enhancer Elements, Genetic ,Cardiovascular and Metabolic Diseases ,Cell culture ,Molecular Probes ,Function and Dysfunction of the Nervous System - Abstract
© 2016 by The American Society of Hematology. Enhancers are the primary determinants of cell identity, and specific promoter/enhancer combinations of Endoglin (ENG) have been shown to target blood and endothelium in the embryo. Here, we generated a series of embryonic stem cell lines, each targeted with reporter constructs driven by specific promoter/enhancer combinations of ENG, to evaluate their discriminative potential and value as molecular probes of the corresponding transcriptome. The Eng promoter (P) in combination with the 28/17/19-kb enhancers, targeted cells in FLK1 mesoderm that were enriched for blast colony forming potential, whereas the P/28-kb enhancer targeted TIE21/c-KIT1/CD412 endothelial cells that were enriched for hematopoieticpotential. These fractions were isolated using reporter expression and their transcriptomes profiled by RNA-seq. There was high concordance between our signatures and those from embryos with defects at corresponding stages of hematopoiesis. Of the 6 genes that were upregulated in both hemogenic mesoderm and hemogenic endothelial fractions targeted by the reporters,LRP2, amultiligandreceptor,wasthe only gene that hadnot previously been associated with hematopoiesis.WeshowthatLRP2is indeedinvolved indefinitivehematopoiesisandbydoingsovalidatetheuseof reportergene-coupled enhancers as probes to gain insights into transcriptional changes that facilitate cell fate transitions.
- Published
- 2016
- Full Text
- View/download PDF
122. Ionic immune suppression within the tumour microenvironment limits T cell effector function
- Author
-
Robert L. Eil, Jenny H. Pan, David S. Schrump, Douglas C. Palmer, Shashank J. Patel, Tori N. Yamamoto, Valentina Carbonaro, Klaus Okkenhaug, Nicholas P. Restifo, David Clever, Rahul Roychoudhuri, Geoffrey Guittard, Zhiya Yu, Alena Gros, Suman K. Vodnala, W. Marston Linehan, Madhusudhanan Sukumar, Christopher A. Klebanoff, Carbonaro, Valentina [0000-0003-0915-6901], Okkenhaug, Klaus [0000-0002-9432-4051], Roychoudhuri, Rahul [0000-0002-5392-1853], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Male ,Potassium Channels ,Potassium ions ,T cell ,medicine.medical_treatment ,T-Lymphocytes ,Receptors, Antigen, T-Cell ,CD8-Positive T-Lymphocytes ,Biology ,Article ,Membrane Potentials ,03 medical and health sciences ,Mice ,Necrosis ,Lymphocytes, Tumor-Infiltrating ,Immune system ,Neoplasms ,medicine ,Immune Tolerance ,Tumor Microenvironment ,Animals ,Humans ,Receptor ,Melanoma ,Multidisciplinary ,Kv1.3 Potassium Channel ,Effector ,TOR Serine-Threonine Kinases ,T-cell receptor ,Immunotherapy ,Cations, Monovalent ,Survival Analysis ,Potassium channel ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Commentary ,Potassium ,Tumor Escape ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Tumours progress despite being infiltrated by tumour-specific effector T cells1. Tumours contain areas of cellular necrosis, which is associated with poor survival in a variety of cancers2. Here, we show that necrosis releases an intracellular ion, potassium, into the extracellular fluid of mouse and human tumours causing profound suppression of T cell effector function. We find that elevations in the extracellular potassium concentration [K+]e act to impair T cell receptor (TCR)-driven Akt-mTOR phosphorylation and effector programmes, this potassium-mediated suppression of Akt-mTOR signalling and T cell function is dependent upon the activity of the serine/threonine phosphatase PP2A3,4. While the suppressive effect mediated by elevated [K+]e is independent of changes in plasma membrane potential (Vm), it does require an increase in intracellular potassium ([K+]i). Concordantly, ionic reprogramming of tumour-specific T cells through overexpression of the potassium channel Kv1.3 lowers [K+]i and improves effector functions in vitro and in vivo. Consequently, Kv1.3 T cell overexpression enhances tumour clearance and survival of melanoma-bearing mice. These results uncover a previously undescribed ionic checkpoint blocking T cell function within tumours and identify new strategies for cancer immunotherapy.
- Published
- 2016
123. Increased cardiovascular mortality more than fifteen years after radiotherapy for breast cancer: a population-based study
- Author
-
David Robinson, Venkata Putcha, Rahul Roychoudhuri, Jack Cuzick, Sarah C. Darby, Henrik Møller, Roychoudhuri, Rahul [0000-0002-5392-1853], and Apollo - University of Cambridge Repository
- Subjects
medicine.medical_specialty ,Cancer Research ,Time Factors ,medicine.medical_treatment ,Breast Neoplasms ,Disease ,lcsh:RC254-282 ,Breast cancer ,Surgical oncology ,Internal medicine ,medicine ,Genetics ,Humans ,Radiation Injuries ,Cardiovascular mortality ,Aged ,business.industry ,Hazard ratio ,Heart ,Middle Aged ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Confidence interval ,Cancer registry ,Surgery ,Radiation therapy ,Oncology ,Cardiovascular Diseases ,Female ,business ,Follow-Up Studies ,Research Article - Abstract
Background Breast radiotherapy as practised in the 1970s and 1980s resulted in significant myocardial exposure, and this was higher when the left breast was treated. It has been proposed that this difference might result in greater cardiovascular mortality following irradiation of the left breast when compared with the right. Methods All cases of female breast cancer diagnosed between 1971 and 1988 and recorded on the Thames Cancer Registry database were followed up to the end of 2003 to identify cases who had died from ischaemic heart disease (IHD) or any cardiovascular disease (CVD). A proportional hazards regression analysis was performed, stratified by time since diagnosis, using as the baseline group those women with right-sided disease who did not receive radiotherapy, and adjusting for age at diagnosis. Results A total of 20,871 women with breast cancer were included in the analysis, of which 51% had left-sided disease. Mortality at 15+ years after diagnosis was increased in recipients of left-breast radiotherapy compared to non-irradiated women with right-sided breast cancer, both for IHD (hazard ratio 1.59; 95% confidence interval 1.21–2.08; p = 0.001) and all CVD (hazard ratio 1.27; 95% confidence interval 1.07–1.51; p = 0.006). When irradiated women with left-sided breast cancer were compared with irradiated women with right-sided breast cancer, cardiovascular mortality at 15+ years after diagnosis was raised by around 25% (IHD: hazard ratio 1.23; 95% confidence interval 0.95–1.60; p = 0.114; CVD: hazard ratio 1.25; 95% confidence interval 1.05–1.49; p = 0.014). Conclusion We have found an elevation in cardiovascular mortality more than 15 years after breast radiotherapy in women diagnosed with breast cancer between 1971 and 1988. The risk was greater following irradiation of the left breast compared with the right. This confirms that radiotherapy as practised in the 1970s and 1980s has resulted in significant long-term cardiac toxicity. In absolute terms, the increase in cardiovascular mortality induced by radiotherapy may be substantial, as these mortality events are relatively common.
- Published
- 2007
124. NaCl enhances CD8 + T cell effector functions in cancer immunotherapy.
- Author
-
Scirgolea C, Sottile R, De Luca M, Susana A, Carnevale S, Puccio S, Ferrari V, Lise V, Contarini G, Scarpa A, Scamardella E, Feno S, Camisaschi C, De Simone G, Basso G, Giuliano D, Mazza EMC, Gattinoni L, Roychoudhuri R, Voulaz E, Di Mitri D, Simonelli M, Losurdo A, Pozzi D, Tsui C, Kallies A, Timo S, Martano G, Barberis E, Manfredi M, Rescigno M, Jaillon S, and Lugli E
- Subjects
- Animals, Mice, Humans, Cell Differentiation, Tumor Microenvironment immunology, Neoplasms immunology, Neoplasms therapy, Neoplasms drug therapy, Cell Line, Tumor, Interferon-gamma metabolism, Glutamine metabolism, Mice, Inbred C57BL, Immunotherapy, Adoptive methods, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Sodium Chloride, Immunotherapy methods
- Abstract
CD8
+ T cells control tumors but inevitably become dysfunctional in the tumor microenvironment. Here, we show that sodium chloride (NaCl) counteracts T cell dysfunction to promote cancer regression. NaCl supplementation during CD8+ T cell culture induced effector differentiation, IFN-γ production and cytotoxicity while maintaining the gene networks responsible for stem-like plasticity. Accordingly, adoptive transfer of tumor-specific T cells resulted in superior anti-tumor immunity in a humanized mouse model. In mice, a high-salt diet reduced the growth of experimental tumors in a CD8+ T cell-dependent manner by inhibiting terminal differentiation and enhancing the effector potency of CD8+ T cells. Mechanistically, NaCl enhanced glutamine consumption, which was critical for transcriptional, epigenetic and functional reprogramming. In humans, CD8+ T cells undergoing antigen recognition in tumors and predicting favorable responses to checkpoint blockade immunotherapy resembled those induced by NaCl. Thus, NaCl metabolism is a regulator of CD8+ T cell effector function, with potential implications for cancer immunotherapy., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
125. IFNγ Production by Functionally Reprogrammed Tregs Promotes Antitumor Efficacy of OX40/CD137 Bispecific Agonist Therapy.
- Author
-
Imianowski CJ, Kuo P, Whiteside SK, von Linde T, Wesolowski AJ, Conti AG, Evans AC, Baird T, Morris BI, Fletcher NE, Yang J, Poon E, Lakins MA, Yamamoto M, Brewis N, Morrow M, and Roychoudhuri R
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Humans, Cell Line, Tumor, Female, T-Lymphocytes, Regulatory drug effects, T-Lymphocytes, Regulatory immunology, Receptors, OX40 agonists, Receptors, OX40 immunology, Tumor Necrosis Factor Receptor Superfamily, Member 9 agonists, Interferon-gamma metabolism, Antibodies, Bispecific pharmacology, Antibodies, Bispecific therapeutic use
- Abstract
Regulatory T cells (Treg) are highly enriched within many tumors and suppress immune responses to cancer. There is intense interest in reprogramming Tregs to contribute to antitumor immunity. OX40 and CD137 are expressed highly on Tregs, activated and memory T cells, and NK cells. In this study, using a novel bispecific antibody targeting mouse OX40 and CD137 (FS120m), we show that OX40/CD137 bispecific agonism induces potent antitumor immunity partially dependent upon IFNγ production by functionally reprogrammed Tregs. Treatment of tumor-bearing animals with OX40/CD137 bispecific agonists reprograms Tregs into both fragile Foxp3+ IFNγ+ Tregs with decreased suppressive function and lineage-instable Foxp3- IFNγ+ ex-Tregs. Treg fragility is partially driven by IFNγ signaling, whereas Treg instability is associated with reduced IL2 responsiveness upon treatment with OX40/CD137 bispecific agonists. Importantly, conditional deletion of Ifng in Foxp3+ Tregs and their progeny partially reverses the antitumor efficacy of OX40/CD137 bispecific agonist therapy, revealing that reprogramming of Tregs into IFNγ-producing cells contributes to the anti-tumor efficacy of OX40/CD137 bispecific agonists. These findings provide insights into mechanisms by which bispecific agonist therapies targeting costimulatory receptors highly expressed by Tregs potentiate antitumor immunity in mouse models., Significance: The bispecific antibody FS120, an immunotherapy currently being tested in the clinic, partially functions by inducing anti-tumor activity of Tregs, which results in tumor rejection., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
126. Cross-talk between ILC2 and Gata3 high T regs locally constrains adaptive type 2 immunity.
- Author
-
Stockis J, Yip T, Moreno-Vicente J, Burton O, Samarakoon Y, Schuijs MJ, Raghunathan S, Garcia C, Luo W, Whiteside SK, Png S, Simpson C, Monk S, Sawle A, Yin K, Barbieri J, Papadopoulos P, Wong H, Rodewald HR, Vyse T, McKenzie ANJ, Cragg MS, Hoare M, Withers DR, Fehling HJ, Roychoudhuri R, Liston A, and Halim TYF
- Subjects
- Animals, Mice, Lymphocytes immunology, Immunity, Innate immunology, Mice, Knockout, Th2 Cells immunology, Female, T-Lymphocytes, Regulatory immunology, GATA3 Transcription Factor immunology, GATA3 Transcription Factor metabolism, Adaptive Immunity immunology, Mice, Inbred C57BL
- Abstract
Regulatory T cells (T
regs ) control adaptive immunity and restrain type 2 inflammation in allergic disease. Interleukin-33 promotes the expansion of tissue-resident Tregs and group 2 innate lymphoid cells (ILC2s); however, how Tregs locally coordinate their function within the inflammatory niche is not understood. Here, we show that ILC2s are critical orchestrators of Treg function. Using spatial, cellular, and molecular profiling of the type 2 inflamed niche, we found that ILC2s and Tregs engage in a direct (OX40L-OX40) and chemotaxis-dependent (CCL1-CCR8) cellular dialogue that enforces the local accumulation of Gata3high Tregs , which are transcriptionally and functionally adapted to the type 2 environment. Genetic interruption of ILC2-Treg communication resulted in uncontrolled type 2 lung inflammation after allergen exposure. Mechanistically, we found that Gata3high Tregs can modulate the local bioavailability of the costimulatory molecule OX40L, which subsequently controlled effector memory T helper 2 cell numbers. Hence, ILC2-Treg interactions represent a critical feedback mechanism to control adaptive type 2 immunity.- Published
- 2024
- Full Text
- View/download PDF
127. Intratumoral antigen signaling traps CD8 + T cells to confine exhaustion to the tumor site.
- Author
-
Takahashi M, So TY, Chamberlain-Evans V, Hughes R, Yam-Puc JC, Kania K, Ruhle M, Mann T, Schuijs MJ, Coupland P, Naisbitt D, Halim TYF, Lyons PA, Lio P, Roychoudhuri R, Okkenhaug K, Adams DJ, Smith KGC, Jodrell DI, Chapman MA, and Thaventhiran JED
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Mice, Transgenic, Antigens, Neoplasm immunology, Neoplasms immunology, CD8-Positive T-Lymphocytes immunology, Signal Transduction immunology
- Abstract
Immunotherapy advances have been hindered by difficulties in tracking the behaviors of lymphocytes after antigen signaling. Here, we assessed the behavior of T cells active within tumors through the development of the antigen receptor signaling reporter (AgRSR) mouse, fate-mapping lymphocytes responding to antigens at specific times and locations. Contrary to reports describing the ready egress of T cells out of the tumor, we find that intratumoral antigen signaling traps CD8
+ T cells in the tumor. These clonal populations expand and become increasingly exhausted over time. By contrast, antigen-signaled regulatory T cell (Treg ) clonal populations readily recirculate out of the tumor. Consequently, intratumoral antigen signaling acts as a gatekeeper to compartmentalize CD8+ T cell responses, even within the same clonotype, thus enabling exhausted T cells to remain confined to a specific tumor tissue site.- Published
- 2024
- Full Text
- View/download PDF
128. GS-TCGA: Gene Set-Based Analysis of The Cancer Genome Atlas.
- Author
-
Baird T and Roychoudhuri R
- Subjects
- Humans, Genome, Gene Expression Regulation, Survival Analysis, Databases, Genetic, Neoplasms genetics
- Abstract
Most tools for analyzing large gene expression datasets, including The Cancer Genome Atlas (TCGA), have focused on analyzing the expression of individual genes or inference of the abundance of specific cell types from whole transcriptome information. While these methods provide useful insights, they can overlook crucial process-based information that may enhance our understanding of cancer biology. In this study, we describe three novel tools incorporated into an online resource; gene set-based analysis of The Cancer Genome Atlas (GS-TCGA). GS-TCGA is designed to enable user-friendly exploration of TCGA data using gene set-based analysis, leveraging gene sets from the Molecular Signatures Database. GS-TCGA includes three unique tools: GS-Surv determines the association between the expression of gene sets and survival in human cancers. Co-correlative gene set enrichment analysis (CC-GSEA) utilizes interpatient heterogeneity in cancer gene expression to infer functions of specific genes based on GSEA of coregulated genes in TCGA. GS-Corr utilizes interpatient heterogeneity in cancer gene expression profiles to identify genes coregulated with the expression of specific gene sets in TCGA. Users are also able to upload custom gene sets for analysis with each tool. These tools empower researchers to perform survival analysis linked to gene set expression, explore the functional implications of gene coexpression, and identify potential gene regulatory mechanisms.
- Published
- 2024
- Full Text
- View/download PDF
129. Tumour-retained activated CCR7 + dendritic cells are heterogeneous and regulate local anti-tumour cytolytic activity.
- Author
-
Lee CYC, Kennedy BC, Richoz N, Dean I, Tuong ZK, Gaspal F, Li Z, Willis C, Hasegawa T, Whiteside SK, Posner DA, Carlesso G, Hammond SA, Dovedi SJ, Roychoudhuri R, Withers DR, and Clatworthy MR
- Subjects
- Humans, Animals, Mice, Receptors, CCR7 genetics, Antigen Presentation, Dendritic Cells, CD8-Positive T-Lymphocytes, Neoplasms genetics, Neoplasms therapy
- Abstract
Tumour dendritic cells (DCs) internalise antigen and upregulate CCR7, which directs their migration to tumour-draining lymph nodes (dLN). CCR7 expression is coupled to an activation programme enriched in regulatory molecule expression, including PD-L1. However, the spatio-temporal dynamics of CCR7
+ DCs in anti-tumour immune responses remain unclear. Here, we use photoconvertible mice to precisely track DC migration. We report that CCR7+ DCs are the dominant DC population that migrate to the dLN, but a subset remains tumour-resident despite CCR7 expression. These tumour-retained CCR7+ DCs are phenotypically and transcriptionally distinct from their dLN counterparts and heterogeneous. Moreover, they progressively downregulate the expression of antigen presentation and pro-inflammatory transcripts with more prolonged tumour dwell-time. Tumour-residing CCR7+ DCs co-localise with PD-1+ CD8+ T cells in human and murine solid tumours, and following anti-PD-L1 treatment, upregulate stimulatory molecules including OX40L, thereby augmenting anti-tumour cytolytic activity. Altogether, these data uncover previously unappreciated heterogeneity in CCR7+ DCs that may underpin a variable capacity to support intratumoural cytotoxic T cells., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
130. Intracellular K+ Limits T-cell Exhaustion and Preserves Antitumor Function.
- Author
-
Collier C, Wucherer K, McWhorter M, Jenkins C, Bartlett A, Roychoudhuri R, and Eil R
- Subjects
- Humans, Animals, Mice, Reactive Oxygen Species metabolism, Sodium-Potassium-Exchanging ATPase genetics, Sodium-Potassium-Exchanging ATPase metabolism, CD8-Positive T-Lymphocytes metabolism, T-Cell Exhaustion, Signal Transduction genetics
- Abstract
T cells are often compromised within cancers, allowing disease progression. We previously found that intratumoral elevations in extracellular K+, related to ongoing cell death, constrained CD8+ T-cell Akt-mTOR signaling and effector function. To alleviate K+-mediated T-cell dysfunction, we pursued genetic means to lower intracellular K+. CD8+ T cells robustly and dynamically express the Na+/K+ ATPase, among other K+ transporters. CRISPR-Cas9-mediated disruption of the Atp1a1 locus lowered intracellular K+ and elevated the resting membrane potential (i.e., Vm, Ψ). Despite compromised Ca2+ influx, Atp1a1-deficient T cells harbored tonic hyperactivity in multiple signal transduction cascades, along with a phenotype of exhaustion in mouse and human CD8+ T cells. Provision of exogenous K+ restored intracellular levels in Atp1a1-deficient T cells and prevented damaging levels of reactive oxygen species (ROS), and both antioxidant treatment and exogenous K+ prevented Atp1a1-deficient T-cell exhaustion in vitro. T cells lacking Atp1a1 had compromised persistence and antitumor activity in a syngeneic model of orthotopic murine melanoma. Translational application of these findings will require balancing the beneficial aspects of intracellular K+ with the ROS-dependent nature of T-cell effector function. See related Spotlight by Banuelos and Borges da Silva, p. 6., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
131. Acquisition of suppressive function by conventional T cells limits antitumor immunity upon T reg depletion.
- Author
-
Whiteside SK, Grant FM, Alvisi G, Clarke J, Tang L, Imianowski CJ, Zhang B, Evans AC, Wesolowski AJ, Conti AG, Yang J, Lauder SN, Clement M, Humphreys IR, Dooley J, Burton O, Liston A, Alloisio M, Voulaz E, Langhorne J, Okkenhaug K, Lugli E, and Roychoudhuri R
- Subjects
- Mice, Humans, Animals, Interleukin-10 metabolism, Immunotherapy, Forkhead Transcription Factors metabolism, T-Lymphocytes, Regulatory, Neoplasms therapy, Neoplasms metabolism
- Abstract
Regulatory T (T
reg ) cells contribute to immune homeostasis but suppress immune responses to cancer. Strategies to disrupt Treg cell-mediated cancer immunosuppression have been met with limited clinical success, but the underlying mechanisms for treatment failure are poorly understood. By modeling Treg cell-targeted immunotherapy in mice, we find that CD4+ Foxp3- conventional T (Tconv ) cells acquire suppressive function upon depletion of Foxp3+ Treg cells, limiting therapeutic efficacy. Foxp3- Tconv cells within tumors adopt a Treg cell-like transcriptional profile upon ablation of Treg cells and acquire the ability to suppress T cell activation and proliferation ex vivo. Suppressive activity is enriched among CD4+ Tconv cells marked by expression of C-C motif receptor 8 (CCR8), which are found in mouse and human tumors. Upon Treg cell depletion, CCR8+ Tconv cells undergo systemic and intratumoral activation and expansion, and mediate IL-10-dependent suppression of antitumor immunity. Consequently, conditional deletion of Il10 within T cells augments antitumor immunity upon Treg cell depletion in mice, and antibody blockade of IL-10 signaling synergizes with Treg cell depletion to overcome treatment resistance. These findings reveal a secondary layer of immunosuppression by Tconv cells released upon therapeutic Treg cell depletion and suggest that broader consideration of suppressive function within the T cell lineage is required for development of effective Treg cell-targeted therapies.- Published
- 2023
- Full Text
- View/download PDF
132. Intracellular K + limits T cell exhaustion and preserves antitumor function.
- Author
-
Collier C, Wucherer K, McWhorter M, Jenkins C, Bartlett A, Roychoudhuri R, and Eil R
- Abstract
The cancer-killing activity of T cells is often compromised within tumors, allowing disease progression. We previously found that intratumoral elevations in extracellular K
+ related to ongoing cell death constrained CD8+ T cell Akt-mTOR signaling and effector function (1,2). To alleviate K+ mediated T cell suppression, we pursued genetic means to lower intracellular K+ . Transcriptomic analysis of CD8+ T cells demonstrated the Na+ /K+ ATPase to be robustly and dynamically expressed. CRISPR-Cas9 mediated deletion of the catalytic alpha subunit of the Na+ /K+ ATPase lowered intracellular K+ but produced tonic hyperactivity in multiple signal transduction cascades along with the acquisition of co-inhibitory receptors and terminal differentiation in mouse and human CD8+ T cells. Mechanistically, Na+ /K+ ATPase disruption led to ROS accumulation due to depletion of intracellular K+ in T cells. Antioxidant treatment or high K+ media prevented Atp1a1 deficient T cells from exhausted T (TEx ) cell formation. Consistent with transcriptional and proteomic data suggesting a TEx cell phenotype, T cells lacking Atp1a1 had compromised persistence and antitumor activity in a syngeneic model of orthotopic murine melanoma. Translational application of these findings will include efforts to lower intracellular K+ while limiting ROS accumulation within tumor specific T cells., Synopsis: High extracellular K+ (↑[K+ ]e ) is found within tumors and suppresses T cell effector function. Collier et al. find that deletion of the Na+ /K+ ATPase in T cells lowers intracellular K+ and promotes ROS accumulation, tonic signal transduction and T cell exhaustion owing to ROS accumulation. Engineering T cell ion transport is an important consideration for cancer immunotherapy.- Published
- 2023
- Full Text
- View/download PDF
133. Orthogonal engineering of synthetic T cell states to enhance cancer immunotherapy.
- Author
-
Conti AG and Roychoudhuri R
- Subjects
- Humans, T-Lymphocytes, Immunotherapy, Adoptive, Genetic Engineering, Immunotherapy, Neoplasms genetics, Neoplasms therapy
- Published
- 2023
- Full Text
- View/download PDF
134. High-Dimensional Single-Cell Profiling of Tumor-Infiltrating CD4 + Regulatory T Cells.
- Author
-
Alvisi G, Puccio S, Roychoudhuri R, Scirgolea C, and Lugli E
- Subjects
- Flow Cytometry, Forkhead Transcription Factors, Humans, Immune Tolerance, Neoplasms, T-Lymphocytes, Regulatory
- Abstract
CD4
+ T regulatory cells (Tregs) are a specialized subset of T lymphocytes, which promote immune homeostasis and tumor immunosuppression by restricting effector T cell immune responses. The characterization of context-specific Treg phenotypic heterogeneity is pivotal to determine their potential contributions to diseases. In the recent years, high-dimensional single-cell technologies, such as single-cell RNA sequencing, mass cytometry, or polychromatic flow cytometry, have played a central role in elucidating the heterogeneity of the Treg compartment at the cellular and molecular levels. Here we describe an example of high-dimensional flow cytometry analysis capable of defining an effector Treg subpopulation that positively correlates with cancer progression. Moreover, we provide a workflow template of high-dimensional single-cell analysis that is readily applicable to any leukocyte subpopulation., (© 2023. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)- Published
- 2023
- Full Text
- View/download PDF
135. Multimodal single-cell profiling of intrahepatic cholangiocarcinoma defines hyperactivated Tregs as a potential therapeutic target.
- Author
-
Alvisi G, Termanini A, Soldani C, Portale F, Carriero R, Pilipow K, Costa G, Polidoro M, Franceschini B, Malenica I, Puccio S, Lise V, Galletti G, Zanon V, Colombo FS, De Simone G, Tufano M, Aghemo A, Di Tommaso L, Peano C, Cibella J, Iannacone M, Roychoudhuri R, Manzo T, Donadon M, Torzilli G, Kunderfranco P, Di Mitri D, Lugli E, and Lleo A
- Subjects
- Humans, Bile Ducts, Intrahepatic pathology, RNA metabolism, T-Lymphocytes, Regulatory, Transcription Factors metabolism, Tumor Microenvironment, Single-Cell Analysis, Bile Duct Neoplasms pathology, Cholangiocarcinoma pathology
- Abstract
Background & Aims: The landscape and function of the immune infiltrate of intrahepatic cholangiocarcinoma (iCCA), a rare, yet aggressive tumor of the biliary tract, remains poorly characterized, limiting development of successful immunotherapies. Herein, we aimed to define the molecular characteristics of tumor-infiltrating leukocytes with a special focus on CD4+ regulatory T cells (Tregs)., Methods: We used high-dimensional single-cell technologies to characterize the T-cell and myeloid compartments of iCCA tissues, comparing these with their tumor-free peritumoral and circulating counterparts. We further used genomics and cellular assays to define the iCCA-specific role of a novel transcription factor, mesenchyme homeobox 1 (MEOX1), in Treg biology., Results: We found poor infiltration of putative tumor-specific CD39+ CD8+ T cells accompanied by abundant infiltration of hyperactivated CD4+ Tregs. Single-cell RNA-sequencing identified an altered network of transcription factors in iCCA-infiltrating compared to peritumoral T cells, suggesting reduced effector functions by tumor-infiltrating CD8+ T cells and enhanced immunosuppression by CD4+ Tregs. Specifically, we found that expression of MEOX1 was highly enriched in tumor-infiltrating Tregs, and demonstrated that MEOX1 overexpression is sufficient to reprogram circulating Tregs to acquire the transcriptional and epigenetic landscape of tumor-infiltrating Tregs. Accordingly, enrichment of the MEOX1-dependent gene program in Tregs was strongly associated with poor prognosis in a large cohort of patients with iCCA., Conclusions: We observed abundant infiltration of hyperactivated CD4+ Tregs in iCCA tumors along with reduced CD8+ T-cell effector functions. Interfering with hyperactivated Tregs should be explored as an approach to enhance antitumor immunity in iCCA., Lay Summary: Immune cells have the potential to slow or halt the progression of tumors. However, some tumors, such as intrahepatic cholangiocarcinoma, are associated with very limited immune responses (and infiltration of cancer-targeting immune cells). Herein, we show that a specific population of regulatory T cells (a type of immune cell that actually suppresses the immune response) are hyperactivated in intrahepatic cholangiocarcinoma. Targeting these cells could enable cancer-targeting immune cells to act more effectively and should be looked at as a potential therapeutic approach to this aggressive cancer type., Competing Interests: Conflict of interest E.L. receives research grants from Bristol Myers Squibb and consulting fees from BD Biosciences. A.L. reports receiving consulting fees from Intercept Pharma, AlfaSigma, Takeda, AbbVie, Gilead, and MSD and travel expenses from Intercept Pharma, AlfaSigma and AbbVie. M.I. participates in advisory boards/consultancies for Gilead Sciences, Roche, Third Rock Ventures, Antios Therapeutics, Amgen, Asher Bio, Allovir, ENYO Pharma. The other authors have no competing interests. Please refer to the accompanying ICMJE disclosure forms for further details., (Copyright © 2022 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
136. Multiply restimulated human thymic regulatory T cells express distinct signature regulatory T-cell transcription factors without evidence of exhaustion.
- Author
-
Hippen KL, Furlan SN, Roychoudhuri R, Wang E, Zhang Y, Osborn MJ, Merkel SC, Hani S, MacMillan ML, Cichocki F, Miller JS, Wagner JE, Restifo NP, Kean LS, and Blazar BR
- Subjects
- Adoptive Transfer, Fetal Blood, Forkhead Transcription Factors genetics, Humans, Graft vs Host Disease, T-Lymphocytes, Regulatory
- Abstract
Background Aims: Adoptive transfer of suppressive CD4+CD25+ thymic regulatory T cells (tTregs) can control auto- and alloimmune responses but typically requires in vitro expansion to reach the target cell number for efficacy. Although the adoptive transfer of expanded tTregs purified from umbilical cord blood ameliorates graft-versus-host disease in patients receiving hematopoietic stem cell transplantation for lymphohematopoietic malignancy, individual Treg products of 100 × 10
6 cells/kg are manufactured over an extended 19-day time period using a process that yields variable products and is both laborious and costly. These limitations could be overcome with the availability of 'off the shelf' Treg., Results: Previously, the authors reported a repetitive restimulation expansion protocol that maintains Treg phenotype (CD4+25++127-Foxp3+), potentially providing hundreds to thousands of patient infusions. However, repetitive stimulation of effector T cells induces a well-defined program of exhaustion that leads to reduced T-cell survival and function. Unexpectedly, the authors found that multiply stimulated human tTregs do not develop an exhaustion signature and instead maintain their Treg gene expression pattern. The authors also found that tTregs expanded with one or two rounds of stimulation and tTregs expanded with three or five rounds of stimulation preferentially express distinct subsets of a group of five transcription factors that lock in Treg Foxp3expression, Treg stability and suppressor function. Multiply restimulated Tregs also had increased transcripts characteristic of T follicular regulatory cells, a Treg subset., Discussion: These data demonstrate that repetitively expanded human tTregs have a Treg-locking transcription factor with stable FoxP3 and without the classical T-cell exhaustion gene expression profile-desirable properties that support the possibility of off-the-shelf Treg therapeutics., Competing Interests: Declaration of Competing Interest BRB is a founder of Tmunity Therapeutics, serves as an advisor for and receives research support from BlueRock Therapeutics and, along with KLH, holds patents for the production and use of Tregs for clinical trials. LSK is on the scientific advisory board for HiFiBio and reports research funding from Bristol Myers Squibb, Kymab Limited, Magenta Therapeutics, BlueBird Bio and Regeneron Pharmaceuticals; consulting fees from Equillium, FortySeven Inc, Novartis Inc, EMD Serono, Gilead Sciences and Takeda Pharmaceuticals; the patent “Method to prevent relapse after transplant,” which is pending; and the patent “Method to prevent GVHD after transplant,” with royalties paid., (Copyright © 2021 International Society for Cell & Gene Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
137. Paths to expansion: Differential requirements of IRF4 in CD8 + T-cell expansion driven by antigen and homeostatic cytokines.
- Author
-
Lugli E, Brummelman J, Pilipow K, and Roychoudhuri R
- Subjects
- Animals, Cell Differentiation immunology, Cell Proliferation, Immunologic Memory immunology, Lymphocyte Activation immunology, Mice, PTEN Phosphohydrolase metabolism, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction immunology, CD8-Positive T-Lymphocytes immunology, Interferon Regulatory Factors immunology, Receptors, Antigen, T-Cell immunology
- Abstract
Interferon regulatory factor 4 (IRF4) regulates the clonal expansion and metabolic activity of activated T cells, but the precise context and mechanisms of its function in these processes are unclear. In this issue of the European Journal of Immunology, Miyakoda et al. [Eur. J. Immunol. 2018. 48: 1319-1328] show that IRF4 is required for activation and expansion of naïve and memory CD8
+ T cells driven by T-cell receptor (TCR) signaling, but dispensable for memory CD8+ T-cell maintenance and homeostatic proliferation driven by homeostatic cytokines. The authors show that the function of IRF4 in CD8+ T-cell expansion is partially dependent upon activation of the PI3K/AKT pathway through direct or indirect attenuation of PTEN expression. These data shed light upon the differential intracellular pathways required for naïve and memory T cells to respond to self-antigens and/or homeostatic cytokines, and highlight the potential translational relevance of these findings in the context of immune reconstitution such as following allogeneic stem cell transplantation., (© 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)- Published
- 2018
- Full Text
- View/download PDF
138. Epigenetic control of CD8 + T cell differentiation.
- Author
-
Henning AN, Roychoudhuri R, and Restifo NP
- Subjects
- Animals, CD8-Positive T-Lymphocytes metabolism, Cellular Reprogramming Techniques methods, Chromatin Assembly and Disassembly genetics, Chromatin Assembly and Disassembly immunology, DNA Methylation immunology, Enhancer Elements, Genetic, Histone Code genetics, Humans, Immunotherapy methods, Models, Genetic, Models, Immunological, Transcription, Genetic, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, Cell Differentiation genetics, Cell Differentiation immunology, Epigenesis, Genetic immunology
- Abstract
Upon stimulation, small numbers of naive CD8
+ T cells proliferate and differentiate into a variety of memory and effector cell types. CD8+ T cells can persist for years and kill tumour cells and virally infected cells. The functional and phenotypic changes that occur during CD8+ T cell differentiation are well characterized, but the epigenetic states that underlie these changes are incompletely understood. Here, we review the epigenetic processes that direct CD8+ T cell differentiation and function. We focus on epigenetic modification of DNA and associated histones at genes and their regulatory elements. We also describe structural changes in chromatin organization that affect gene expression. Finally, we examine the translational potential of epigenetic interventions to improve CD8+ T cell function in individuals with chronic infections and cancer.- Published
- 2018
- Full Text
- View/download PDF
139. Lineage relationship of CD8(+) T cell subsets is revealed by progressive changes in the epigenetic landscape.
- Author
-
Crompton JG, Narayanan M, Cuddapah S, Roychoudhuri R, Ji Y, Yang W, Patel SJ, Sukumar M, Palmer DC, Peng W, Wang E, Marincola FM, Klebanoff CA, Zhao K, Tsang JS, Gattinoni L, and Restifo NP
- Subjects
- Animals, Chromatin Assembly and Disassembly genetics, Enhancer Elements, Genetic genetics, Gene Expression Profiling, Histones metabolism, Immunologic Memory genetics, Lymphocyte Subsets metabolism, Methylation, Mice, Inbred C57BL, Promoter Regions, Genetic genetics, Protein Processing, Post-Translational, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes metabolism, Cell Lineage genetics, Cell Lineage immunology, Epigenesis, Genetic
- Abstract
To better elucidate epigenetic mechanisms that correlate with the dynamic gene expression program observed upon T-cell differentiation, we investigated the genomic landscape of histone modifications in naive and memory CD8(+) T cells. Using a ChIP-Seq approach coupled with global gene expression profiling, we generated genome-wide histone H3 lysine 4 (H3K4me3) and H3 lysine 27 (H3K27me3) trimethylation maps in naive, T memory stem cells, central memory cells, and effector memory cells in order to gain insight into how histone architecture is remodeled during T cell differentiation. We show that H3K4me3 histone modifications are associated with activation of genes, while H3K27me3 is negatively correlated with gene expression at canonical loci and enhancers associated with T-cell metabolism, effector function, and memory. Our results also reveal histone modifications and gene expression signatures that distinguish the recently identified T memory stem cells from other CD8(+) T-cell subsets. Taken together, our results suggest that CD8(+) lymphocytes undergo chromatin remodeling in a progressive fashion. These findings have major implications for our understanding of peripheral T-cell ontogeny and the formation of immunological memory.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.