28 results on '"Sasikanth Manne"'
Search Results
2. PD-1 directed immunotherapy alters Tfh and humoral immune responses to seasonal influenza vaccine
- Author
-
Ramin Sedaghat Herati, David A. Knorr, Laura A. Vella, Luisa Victoria Silva, Lakshmi Chilukuri, Sokratis A. Apostolidis, Alexander C. Huang, Alexander Muselman, Sasikanth Manne, Oliva Kuthuru, Ryan P. Staupe, Sharon A. Adamski, Senthil Kannan, Raj K. Kurupati, Hildegund C. J. Ertl, Jeffrey L. Wong, Stylianos Bournazos, Suzanne McGettigan, Lynn M. Schuchter, Ritesh R. Kotecha, Samuel A. Funt, Martin H. Voss, Robert J. Motzer, Chung-Han Lee, Dean F. Bajorin, Tara C. Mitchell, Jeffrey V. Ravetch, and E. John Wherry
- Subjects
Adult ,Influenza Vaccines ,Vaccination ,Immunology ,Humans ,Immunology and Allergy ,Seasons ,T-Lymphocytes, Helper-Inducer ,Immunity, Humoral - Abstract
Anti-programmed death-1 (anti-PD-1) immunotherapy reinvigorates CD8 T cell responses in patients with cancer but PD-1 is also expressed by other immune cells, including follicular helper CD4 T cells (Tfh) which are involved in germinal centre responses. Little is known, however, about the effects of anti-PD-1 immunotherapy on noncancer immune responses in humans. To investigate this question, we examined the impact of anti-PD-1 immunotherapy on the Tfh-B cell axis responding to unrelated viral antigens. Following influenza vaccination, a subset of adults receiving anti-PD-1 had more robust circulating Tfh responses than adults not receiving immunotherapy. PD-1 pathway blockade resulted in transcriptional signatures of increased cellular proliferation in circulating Tfh and responding B cells compared with controls. These latter observations suggest an underlying change in the Tfh-B cell and germinal centre axis in a subset of immunotherapy patients. Together, these results demonstrate dynamic effects of anti-PD-1 therapy on influenza vaccine responses and highlight analytical vaccination as an approach that may reveal underlying immune predisposition to adverse events.
- Published
- 2022
3. Autoreactive CD8+ T cells are restrained by an exhaustion-like program that is maintained by LAG3
- Author
-
Stephanie Grebinoski, Qianxia Zhang, Anthony R. Cillo, Sasikanth Manne, Hanxi Xiao, Erin A. Brunazzi, Tracy Tabib, Carly Cardello, Christine G. Lian, George F. Murphy, Robert Lafyatis, E. John Wherry, Jishnu Das, Creg J. Workman, and Dario A. A. Vignali
- Subjects
Immunology ,Immunology and Allergy - Published
- 2022
4. Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation
- Author
-
Mohamed S. Abdel-Hakeem, Mohammed-Alkhatim Ali, Allison R. Greenplate, Sasikanth Manne, Golnaz Vahedi, Zeyu Chen, Jean-Christophe Beltra, E. John Wherry, Josephine R. Giles, Divij Mathew, Erietta Stelekati, Kito Nzingha, and John L. Johnson
- Subjects
epigenetic landscape of exhausted T cells ,Transcription, Genetic ,T cell ,Immunology ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Lymphocytic Choriomeningitis ,Biology ,Article ,Cell Line ,Epigenesis, Genetic ,Mice ,Immune system ,Antigen ,Cricetinae ,Chlorocebus aethiops ,medicine ,Animals ,Lymphocytic choriomeningitis virus ,Immunology and Allergy ,Cytotoxic T cell ,Hepatocyte Nuclear Factor 1-alpha ,Epigenetics ,recall capacity ,Antigens, Viral ,Vero Cells ,Epigenomics ,T cell exhaustion ,Cell Differentiation ,Acquired immune system ,Chromatin ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,immunological recovery ,Female ,Immunologic Memory - Abstract
Exhausted CD8 T cells (TEX) are a distinct state of T cell differentiation associated with failure to clear chronic viruses and cancer. Immunotherapies such as PD-1 blockade can reinvigorate TEX cells, but reinvigoration is not durable. A major unanswered question is whether TEX cells differentiate into functional durable memory T cells (TMEM) upon antigen clearance. Here, using a mouse model, we found that upon eliminating chronic antigenic stimulation, TEX cells partially (re)acquire phenotypic and transcriptional features of TMEM cells. These ‘recovering’ TEX cells originated from the T cell factor (TCF-1+) TEX progenitor subset. Nevertheless, the recall capacity of these recovering TEX cells remained compromised as compared to TMEM cells. Chromatin-accessibility profiling revealed a failure to recover core memory epigenetic circuits and maintenance of a largely exhausted open chromatin landscape. Thus, despite some phenotypic and transcriptional recovery upon antigen clearance, exhaustion leaves durable epigenetic scars constraining future immune responses. These results support epigenetic remodeling interventions for TEX cell–targeted immunotherapies. Wherry and colleagues examine whether exhausted T cells (TEX) can differentiate into functional memory T cells (TMEM) when chronic antigen is withdrawn. Using the chronic LCMV infection mouse model, they show that ‘recovering’ TEX cells (REC-TEX) only partially recover immunophenotypic and functional characteristics of TMEM cells. The epigenomic status of REC-TEX cells more closely resembles that of TEX cells, and, upon rechallenge, the REC-TEX cells were still compromised in their ability to respond to virus.
- Published
- 2021
5. Neuropilin-1 is a T cell memory checkpoint limiting long-term antitumor immunity
- Author
-
Robert L. Ferris, E. John Wherry, Andrea L. Szymczak-Workman, Ellen N. Scott, Evan J. Lipson, Creg J. Workman, Sasikanth Manne, Dario A. A. Vignali, Ashwin Somasundaram, Angela M. Gocher, Daniel P. Normolle, Tullia C. Bruno, Kate M. Vignali, and Chang Liu
- Subjects
0301 basic medicine ,T cell ,medicine.medical_treatment ,Immunology ,T-cell receptor ,Immunotherapy ,Biology ,Immune checkpoint ,Immune tolerance ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,Memory cell ,medicine ,Cancer research ,Immunology and Allergy ,CD8 ,030215 immunology - Abstract
Robust CD8+ T cell memory is essential for long-term protective immunity but is often compromised in cancer, where T cell exhaustion leads to loss of memory precursors. Immunotherapy via checkpoint blockade may not effectively reverse this defect, potentially underlying disease relapse. Here we report that mice with a CD8+ T cell–restricted neuropilin-1 (NRP1) deletion exhibited substantially enhanced protection from tumor rechallenge and sensitivity to anti-PD1 immunotherapy, despite unchanged primary tumor growth. Mechanistically, NRP1 cell-intrinsically limited the self-renewal of the CD44+PD1+TCF1+TIM3− progenitor exhausted T cells, which was associated with their reduced ability to induce c-Jun/AP-1 expression on T cell receptor restimulation, a mechanism that may contribute to terminal T cell exhaustion at the cost of memory differentiation in wild-type tumor-bearing hosts. These data indicate that blockade of NRP1, a unique ‘immune memory checkpoint’, may promote the development of long-lived tumor-specific Tmem that are essential for durable antitumor immunity. T cell exhaustion limits antitumor immune responses. Vignali and colleagues identify neuropilin-1 as a novel immune checkpoint that cell-intrinsically operates to limit memory cell formation and can be targeted to enhance antitumor responses.
- Published
- 2020
6. Human epigenetic and transcriptional T cell differentiation atlas for identifying functional T cell-specific enhancers
- Author
-
Josephine R. Giles, Sasikanth Manne, Elizabeth Freilich, Derek A. Oldridge, Amy E. Baxter, Sangeeth George, Zeyu Chen, Hua Huang, Lakshmi Chilukuri, Mary Carberry, Lydia Giles, Nan-Ping P. Weng, Regina M. Young, Carl H. June, Lynn M. Schuchter, Ravi K. Amaravadi, Xiaowei Xu, Giorgos C. Karakousis, Tara C. Mitchell, Alexander C. Huang, Junwei Shi, and E. John Wherry
- Subjects
Epigenomics ,Infectious Diseases ,Immunology ,Immunology and Allergy ,Humans ,Cell Differentiation ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Chromatin ,Article ,Epigenesis, Genetic - Abstract
The clinical benefit of T cell immunotherapies remains limited by incomplete understanding of T cell differentiation and dysfunction. We generated an epigenetic and transcriptional atlas of T cell differentiation from healthy humans that included exhausted CD8 T cells and applied this resource in three ways. First, we identified modules of gene expression and chromatin accessibility, revealing molecular coordination of differentiation after activation and between central memory and effector memory. Second, we applied this healthy molecular framework to three settings - a neoadjuvant anti-PD1 melanoma trial, a basal cell carcinoma scATAC-seq data set, and autoimmune disease-associated SNPs - yielding insights into disease-specific biology. Third, we predicted genome-wide cis-regulatory elements and validated this approach for key effector genes using CRISPR interference, providing functional annotation and demonstrating the ability to identify targets for non-coding cellular engineering. These studies define epigenetic and transcriptional regulation of human T cells and illustrate the utility of interrogating disease in the context of a healthy T cell atlas.
- Published
- 2021
7. Signaling through FcγRIIA and the C5a-C5aR pathway mediates platelet hyperactivation in COVID-19
- Author
-
Jeanette Dougherty, Sasikanth Manne, Amy E. Baxter, Josephine R. Giles, Yinghui Jane Huang, Ajinkya Pattekar, Oliva Kuthuru, Jennifer E. Wu, Aae Suzuki, Nuala J. Meyer, Debora Dunbar, Zeyu Chen, Alexander C. Huang, Ariel R. Weisman, Allison R. Greenplate, Mortimer Poncz, Charles S. Abrams, Ian Frank, Liang Zhao, Caroline A. G. Ittner, Mohamed S. Abdel-Hakeem, Amrita Sarkar, Rishi R. Goel, Sigrid Gouma, Divij Mathew, Sokratis A. Apostolidis, John P. Reilly, Derek A. Oldridge, Cécile Alanio, Scott E. Hensley, Laura A. Vella, E. John Wherry, Heather M. Giannini, Lubica Rauova, and Brittany Weiderhold
- Subjects
Adult ,Blood Platelets ,Male ,Morpholines ,Immunology ,Aminopyridines ,Syk ,Complement C5a ,Inflammation ,Fostamatinib ,Severity of Illness Index ,Article ,Thromboembolism ,medicine ,Humans ,Syk Kinase ,Immunology and Allergy ,Platelet ,Platelet activation ,Receptor, Anaphylatoxin C5a ,Cells, Cultured ,biology ,Hyperactivation ,SARS-CoV-2 ,business.industry ,Receptors, IgG ,COVID-19 ,Platelet Activation ,Hospitalization ,Ferritin ,Pyrimidines ,biology.protein ,Female ,medicine.symptom ,Signal transduction ,business ,Signal Transduction ,medicine.drug - Abstract
Patients with COVID-19 present with a wide variety of clinical manifestations. Thromboembolic events constitute a significant cause of morbidity and mortality in patients infected with SARS-CoV-2. Severe COVID-19 has been associated with hyperinflammation and pre-existing cardiovascular disease. Platelets are important mediators and sensors of inflammation and are directly affected by cardiovascular stressors. In this report, we found that platelets from severely ill, hospitalized COVID-19 patients exhibit higher basal levels of activation measured by P-selectin surface expression, and have a poor functional reserve upon in vitro stimulation. Correlating clinical features to the ability of plasma from COVID-19 patients to stimulate control platelets identified ferritin as a pivotal clinical marker associated with platelet hyperactivation. The COVID-19 plasma-mediated effect on control platelets was highest for patients that subsequently developed inpatient thrombotic events. Proteomic analysis of plasma from COVID-19 patients identified key mediators of inflammation and cardiovascular disease that positively correlated with in vitro platelet activation. Mechanistically, blocking the signaling of the FcγRIIa-Syk and C5a-C5aR pathways on platelets, using antibody-mediated neutralization, IgG depletion or the Syk inhibitor fostamatinib, reversed this hyperactivity driven by COVID-19 plasma and prevented platelet aggregation in endothelial microfluidic chamber conditions, thus identifying these potentially actionable pathways as central for platelet activation and/or vascular complications in COVID-19 patients. In conclusion, we reveal a key role of platelet-mediated immunothrombosis in COVID-19 and identify distinct, clinically relevant, targetable signaling pathways that mediate this effect. These studies have implications for the role of platelet hyperactivation in complications associated with SARS-CoV-2 infection., Cover illustration, One-sentence summary: The FcγRIIA and C5a-C5aR pathways mediate platelet hyperactivation in COVID-19
- Published
- 2021
8. PD-1 restricts the development and effector function of tissue regulatory T cells in experimental autoimmune encephalomyelitis
- Author
-
Dan Liang, Jennifer Judge, Samuel Markson, Samantha Guinn, Jenna Lynn Collier, Osmaan Shahid, Sasikanth Manne, Juhi Kuchroo, Megan Fung, Kristen E Pauken, Dario AA Vignali, John E Wherry, and Arlene H Sharpe
- Subjects
Immunology ,Immunology and Allergy - Abstract
PD-1 restricts T cell effector functions, regulates T cell tolerance, and maintains immune homeostasis, thereby playing pivotal roles in cancer, autoimmune and infectious diseases. Our studies and others show that PD-1 can restrain Treg suppressive function in autoimmunity and cancer. Here, we investigate the role of Treg intrinsic PD-1 in regulating pathogenic and protective CD4+ T cell responses in experimental autoimmune encephalomyelitis (EAE) by inducing PD-1 deletion on FoxP3+ cells prior to disease development. This deletion of PD-1 only in Treg protects mice from severe disease, and both flow cytometry and transcriptional analysis of central nervous system (CNS) CD4+ T cells show enrichment of tissue effector Treg (eTreg) subsets at peak of disease. We find that PD-1 restrains the generation and suppressive function of these effector Treg. This tissue eTreg signature is also observed in human and mouse Treg subsets from the tumor microenvironment, and are potent suppressor populations. Further studies are underway to determine how PD-1 impacts TCR signaling and FoxP3 transcriptional co-activators in Treg cells. Our study provides deeper insights into how PD-1 regulates T cell tolerance, underscoring the important role of PD-1 in regulating Treg differentiation and suppressive function during autoimmune diseases, and provide mechanistic insights for PD-1 modulation in cancer and autoimmunity. Supported by NIH P01AI108545
- Published
- 2022
9. 253 PD1 and LAG3 converge to limit polyfunctionality and systemic immunity
- Author
-
Dario A. A. Vignali, E. John Wherry, Creg J. Workman, Lawrence C. Andrews, and Sasikanth Manne
- Subjects
Pharmacology ,Physics ,Cancer Research ,LAG3 ,Oncology ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Molecular Medicine ,Immunology and Allergy ,Systemic immunity ,Limit (mathematics) ,RC254-282 - Abstract
BackgroundTargeting PD1 with monoclonal antibodies has yielded clinical success across a variety of tumor types, yet overcoming inhibitory receptor (IR)-mediated tolerance is essential to improve immunotherapeutic responses. LAG3 co-expresses with PD1 on CD8+ tumor-infiltrating T cells (TIL), signifying a highly exhausted phenotype, and dual PD1/LAG3 blockade in C57BL/6 mice enhances antitumor immunity. As CD8+ TIL is the dominant LAG3-expressing TIL population, it is hypothesized that PD1 and LAG3 synergizes to limit CD8+ TIL function controlling antitumor immunity.MethodsTo understand the cellular and mechanistic basis for PD1/LAG3 synergy, conditional knock-in mice ”surgically dissect” Pdcd1 and/or Lag3 floxed alleles restricted to CD8+ T cells expressing E8ICre.GFP. These mice were crossed with the Pmel transgene to assess PD1 and/or LAG3-sufficient or deficient gp100-specific CD8+ T cell populations. CD8+ Pmel cells were isolated and adoptively transferred into C57BL/6 mice harboring a B16-gp100-overexpressing tumor to observe therapeutic benefit, or to assess T cell functionality within the tumor.ResultsWhile little therapeutic benefit was observed with a prophylactic adoptive transfer of wild-type CD8+ Pmel cells into mice which then received B16-gp100 tumor, there was reduced tumor growth in mice receiving PD1-deficient CD8+ Pmel cells, which was further enhanced in mice receiving PD1/LAG3-deficient CD8+ Pmel cells with long-term tumor-free survival. Likewise, a therapeutic administration of PD1/LAG3-deficient CD8+ Pmel cells into mice once the tumor has been established showed an initial therapeutic benefit, with enhanced survival, that was not demonstrated with adoptive transfer of PD1 or LAG3-deficient, or wild-type, counterparts. Each PD1 and/or LAG3-sufficient or deficient CD8+ Pmel mice were differentially congenically marked to assess each of the four genotypes that can be adoptively transferred into the same host as a ”quad transfer” system. Recovery of these populations within the tumor show that the PD1/LAG3-deficient CD8+ Pmel cells out-compete PD1 or LAG3-deficient, or wild-type, counterparts due to enhanced proliferation (Ki67/BrdU). Furthermore, PD1/LAG3-deficient CD8+ T cells were more functional with increased IFNg and GzmB release observed by flow cytometry.ConclusionsOverall PD1 and LAG3 limit anti-tumor immune effects as removal of both IRs on a gp100 antigen-specific CD8+ T cell population results in reduced B16-gp100 tumor growth and enhanced survival in an adoptive transfer model, as a result of enhanced CD8+ TIL functionality and proliferation. These results provide striking evidence that the development of anti-LAG3 agents in the clinic would yield improved responses with anti-PD1.
- Published
- 2021
10. 310 T cell intrinsic DNA damage and repair response as a novel marker associated with clinical response to PD-1 blockade
- Author
-
Ramin S. Herati, Claire F. Friedman, Divij Mathew, Yuki Muroyama, Caiyue Xu, Alexander C. Huang, Allison R. Greenplate, E. John Wherry, D. Zamarin, Lakshmi Chilukuri, Derek A. Oldridge, and Sasikanth Manne
- Subjects
Pharmacology ,Cancer Research ,Chemistry ,DNA damage ,T cell ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,body regions ,medicine.anatomical_structure ,Oncology ,Cancer research ,medicine ,Molecular Medicine ,Immunology and Allergy ,Pd 1 blockade ,RC254-282 - Abstract
BackgroundDespite the success of immune checkpoint blockade (ICB), many patients still fail to achieve durable clinical benefit. Previous studies have shown that CD8 T cells are reinvigorated by ICB. However, not all patients with this immunological response experience an effective clinical response, suggesting additional parameters may be relevant.DNA damage and repair (DDR) has been extensively studied in the context of inducing cell death of highly-proliferating tumor cells. However, whether T cell-intrinsic DDR impacts T cell differentiation and function, and how the coordination of DDR affects immunological and clinical response to proliferation-inducing ICBs have been largely unexplored. We hypothesized that the T celI-intrinsic DDR responses to proliferative and genotoxic stress might contribute to the disparity between immunological and clinical response.MethodsTo understand the impact of cell-intrinsic DDR on T cell differentiation and responses to cancer therapies, we developed a novel high-dimensional cytometry platform. This DDR-Immune platform enables simultaneous analysis of T cell differentiation state and multiple DDR pathways at single cell resolution. We then investigated immune reinvigoration and its association with DDR, in a cohort of chemotherapy-resistant hypermutated or microsatellite instability-high (MSI-H) uterine cancer patients treated with nivolumab. Peripheral blood samples were examined every 2–4 weeks after initiating anti-PD-1 treatment (N = 21).ResultsThe DDR-Immune platform revealed consistent T cell subset specific patterns of DDR, as well as specific DDR pathways induced by different types of DNA damage, such as γ-irradiation (IR), UV irradiation (UV) or proliferative stress (i.e. anti-CD3/CD28 stimulation). For example, terminally differentiated effector cells had higher DNA damage accumulation and cell death. In contrast, stem cell memory (TSCM) and regulatory T cells (Treg) displayed high DDR with less cell death, suggesting better cell-intrinsic DDR against genotoxic stress for survival advantage. In hypermutated MSI-H uterine cancer patients, CD8 T cells underwent rapid pharmacodynamic proliferation 2–4 weeks after starting PD-1 blockade, which did not correlate with clinical response. Application of the DDR-Immune platform to this cohort revealed, however, that in clinical responders but not clinical non-responders, Ki67+ CD8 T cells responding to PD-1 blockade had rapid induction of DDR represented as a spike increase of phosphorylated-ATM, presumably adapting T cell ‘fitness’ in response to proliferative stress induced by PD-1 blockade.ConclusionsCollectively, the new platform reveals previously unrecognized roles for T cell-intrinsic DDR as a novel determinant of immune responsiveness and clinical outcome to ICB and have potential application to other cancer therapies including chemotherapy and radiotherapy.Ethics ApprovalThe study was approved by MSKCC Ethics Board, approval number 17–180 (NCT03241745).
- Published
- 2021
11. Author Correction: Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation
- Author
-
Sasikanth Manne, Golnaz Vahedi, Allison R. Greenplate, Erietta Stelekati, Josephine R. Giles, Zeyu Chen, E. John Wherry, Divij Mathew, Mohamed S. Abdel-Hakeem, Mohammed-Alkhatim Ali, Kito Nzingha, Jean-Christophe Beltra, and John L. Johnson
- Subjects
Antigenic stimulation ,Text mining ,business.industry ,Immunology ,Immunology and Allergy ,Medicine ,Epigenetics ,Bioinformatics ,business - Published
- 2021
12. TCF-1-centered transcriptional network drives an effector versus exhausted CD8 T cell fate decision
- Author
-
Zeyu Chen, Jean Christophe Beltra, Sasikanth Manne, Golnaz Vahedi, Caiyue Xu, Amy E. Baxter, Sixiang Yu, Chi Wai Lau, Josephine R. Giles, Erietta Stelekati, John L. Johnson, Zhicheng Ji, Omar Khan, Bertram Bengsch, Jennifer E. Wu, Alexander C. Huang, Laura A. Vella, Hongkai Ji, Ramin S. Herati, Shin Foong Ngiow, E. John Wherry, Ryan P. Staupe, Laura M. McLane, Shelley L. Berger, Xiaolu Yang, Makoto Kurachi, and Zhangying Cai
- Subjects
0301 basic medicine ,animal structures ,Transcription, Genetic ,Immunology ,Population ,Programmed Cell Death 1 Receptor ,Biology ,CD8-Positive T-Lymphocytes ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Transcription (biology) ,Precursor cell ,T Cell Transcription Factor 1 ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Gene Regulatory Networks ,education ,Transcription factor ,Progenitor ,education.field_of_study ,Effector ,Gene Expression Profiling ,Cell Differentiation ,Cell biology ,Gene expression profiling ,030104 developmental biology ,Infectious Diseases ,Virus Diseases ,030220 oncology & carcinogenesis ,Chronic Disease ,Host-Pathogen Interactions ,embryonic structures - Abstract
TCF-1 is a key transcription factor in progenitor exhausted CD8 T cells (Tex). Moreover, this Tex cell subset mediates responses to PD-1 checkpoint pathway blockade. However, the role of the transcription factor TCF-1 in early fate decisions and initial generation of Tex cells is unclear. Single cell RNA-sequencing (scRNA-seq) and lineage tracing identified a TCF-1(+)Ly108(+)PD-1(+) CD8 T cell population early during chronic infection that seeds development of mature Tex cells. TCF-1 mediated the bifurcation between divergent fates, repressing development of terminal KLRG1(Hi) effectors while fostering KLRG1(Lo) Tex precursor cells, and PD-1 stabilized this TCF-1(+) Tex precursor cell pool. TCF-1 mediated a T-bet to Eomes transcription factor transition in Tex precursors by promoting Eomes expression and drove c-Myb expression that controlled Bcl-2 and survival. These data define a role for TCF-1 in early fate bifurcation driving Tex precursor cells, and also identify PD-1 as a protector of this early TCF-1 subset.
- Published
- 2019
13. Therapeutic Targeting of Vasculature in the Premetastatic and Metastatic Niches Reduces Lung Metastasis
- Author
-
Jalpa Patel, Surya Kumari Vadrevu, Sanjay K. Srivastava, Sasikanth Manne, Shanawaz M. Ghouse, Magdalena Karbowniczek, Niraj Lodhi, Ashok Silwal, Yvonne Paterson, Maciej M. Markiewski, Bhaumik Patel, and Britney Reese
- Subjects
Lung Neoplasms ,Angiogenesis ,medicine.medical_treatment ,Immunology ,Angiogenesis Inhibitors ,Complement C5a ,Cancer Vaccines ,C5a receptor ,Article ,Metastasis ,Neovascularization ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Tumor Microenvironment ,Immunology and Allergy ,Animals ,Humans ,Neoplasm Metastasis ,Lung ,Receptor, Anaphylatoxin C5a ,Mice, Knockout ,Tumor microenvironment ,Neovascularization, Pathologic ,business.industry ,Myeloid-Derived Suppressor Cells ,Mammary Neoplasms, Experimental ,Immunotherapy ,medicine.disease ,Combined Modality Therapy ,Listeria monocytogenes ,Matrix Metalloproteinase 9 ,Myeloid-derived Suppressor Cell ,Cancer research ,Female ,medicine.symptom ,business ,030215 immunology - Abstract
In the metastasis-targeted organs, angiogenesis is essential for the progression of dormant micrometastases to rapidly growing and clinically overt lesions. However, we observed changes suggesting angiogenic switching in the mouse lungs prior to arrival of tumor cells (i.e., in the premetastatic niche) in the models of breast carcinoma. This angiogenic switching appears to be caused by myeloid-derived suppressor cells recruited to the premetastatic lungs through complement C5a receptor 1 signaling. These myeloid cells are known to secrete several proangiogenic factors in tumors, including IL-1β and matrix metalloproteinase-9, and we found upregulation of these genes in the premetastatic lungs. Blockade of C5a receptor 1 synergized with antiangiogenic Listeria monocytogenes–based vaccines to decrease the lung metastatic burden by reducing vascular density and improving antitumor immunity in the lungs. This was mediated even when growth of primary breast tumors was not affected by these treatments. This work provides initial evidence that angiogenesis contributes to the premetastatic niche in rapidly progressing cancers and that inhibiting this process through immunotherapy is beneficial for reducing or even preventing metastasis.
- Published
- 2019
14. TET2 controls differentiation of terminally exhausted CD8 T cells
- Author
-
Martha S Jordan, Sydney Drury, Josephine R Giles, Sasikanth Manne, Hua Huang, Zeyu Chen, Derek Oldridge, E. John Wherry, and Amy E Baxter
- Subjects
Immunology ,Immunology and Allergy - Abstract
CD8 T cells are critical for clearing infections and immune surveillance of cancers. However, in the setting of chronic viral infections and tumor growth, CD8 T cells become exhausted. T cell exhaustion (TEX) is characterized by a progressive increase of inhibitory receptor expression and concomitant diminishment of effector function. This diminished function renders the host unable to clear the virus or cancer. The TEX population is heterogenous and can be divided broadly into TEX progenitor and TEX terminal populations. TEX progenitor are self-renewing, responsive to PD-1 blockade and give rise to the later TEX subsets, including short-lived TEX terminal cells. Control of DNA demethylation is required in multiple settings for maintenance or loss of stem-like characteristics. We report that TET2, a methylcytosine dioxygenase that mediates active DNA demethylation, is required for TEX progenitors to differentiate fully into TEX terminal cells. TET2-deficiency during persistent viral infection resulted in a numerical loss of TEX terminal cells, while maintaining the TEX progenitor population. The TEX terminal cells that escaped this differentiation block retained some features of TEX progenitors, including decreased expression of several inhibitory receptors. TET2-deficient TEX progenitors responded to PD-1 blockade but were unable to establish expansion of TEX cells thought to be required for viral control. These data highlight the role of DNA methylation and TET2 in establishing TEX diversity.
- Published
- 2021
15. A role for the transcription factor STAT5 in antagonizing CD8+ T cell exhaustion
- Author
-
Jean-Christophe Beltra, Sasikanth Manne, Mohamed S Abdel-Hakeem, Kito Nzingha, Zhen Zhang, Hua Huang, Makoto Kurachi, Yuki Muroyama, Yinghui Jane Huang, Leon L Su, Lora Picton, Helene Decaluwe, Alexander C Huang, Shelley L Berger, Christopher K Garcia, and E. John Wherry
- Subjects
Immunology ,Immunology and Allergy - Abstract
Exhaustion gradually establishes in chronically stimulated CD8s and this process is not reverted by current therapeutic approaches due to establishment of a stable epigenetic program. Recent advances have informed on the developmental process of exhaustion and highlighted TOX as a key lineage-defining TF in the process. Yet, little remains known on molecular pathways capable of antagonizing the TOX-dependent exhaustion program. By depicting transcriptional changes at key developmental steps of exhaustion, we demonstrate an antagonistic role for the TF STAT5 in the development of CD8 T cell exhaustion. STAT5 transcriptional network is heavily silenced upon chronic antigenic stimulation in a TOX-dependent manner which allows initiation of the exhaustion lineage. Increasing STAT5 activity abrogates establishment of the exhaustion lineage leading to the development of effector-like CD8s that acquire a unique transcriptional identity, distinct from exhausted cells, persist throughout chronicity and demonstrate higher protective capacity. Using temporal loss and gain of function approaches, we show that STAT5 triggers loss of progenitor identity by exhausted CD8s (Tex) and subsequent differentiation into the recently identified effector-like intermediate Tex subset. Temporal increase in STAT5 activity also robustly synergizes with PD-L1 blockade by further fostering intermediate Tex cells accumulation. Together, we show that modulating STAT5 activity may counteract the exhaustion process and favor instigation of effector-like characteristic in Tex cells suitable for optimal therapeutic efficacy. This work is supported by the Parker Institute for Cancer Immunotherapy (PICI). JC-Beltra is a PICI scholar awardee.
- Published
- 2021
16. T cell intrinsic DNA damage and repair response as a novel marker associated with clinical response to PD-1 blockade
- Author
-
Yuki Muroyama, Alexander C. Huang, Sasikanth Manne, Ramin S. Herati, Divij Mathew, Caiyue Xu, Lakshmi Chilukuri, Dmitriy Zamarin, Claire F Friedman, and E John Wherry
- Subjects
Immunology ,Immunology and Allergy - Abstract
Despite the high tumor mutational burden (TMB), immune checkpoint blockade (ICB) still fails in some microsatellite instability-high (MSI-H) tumors, suggesting the underlying T cell dysfunction. Immune profiling of PBMC from chemotherapy-resistant MSI-H uterine cancer patients treated with nivolumab identified the proliferative T cell response 2–4 weeks post PD-1 blockade. However, this “immunological response” to ICB was observed regardless of clinical response, suggesting the additional determinants of clinical outcome beyond the proliferative response or high TMB. To test the hypothesis that the gap between immunological and clinical response to ICB was related to T celI-intrinsic response to genotoxic and/or proliferative stress, a novel high dimensional cytometry platform was developed. The platform enables simultaneous analysis of T cell differentiation state and interrogation of multiple DNA damage and repair response (DDR) pathways at single cell resolution. This DDR-Immune platform revealed consistent T cell subset specific patterns of DDR, as well as specific DDR pathways induced by different types of DNA damage. Application of the DDR-Immune platform to this cohort revealed that, in clinical responders, Ki67+ CD8 T cells had rapid increase of pATM, presumably in response to proliferative stress induced by PD-1 blockade. This DDR was not clearly observed in clinical non-responders. In addition, Tregs from clinical non-responders had elevated DDRs compared to responders, suggesting their greater resistance to genotoxic/proliferative stress. Collectively, the new platform reveals previously unrecognized roles for T cell-intrinsic DDR as a novel determinant of immune responsiveness and clinical outcome.
- Published
- 2020
17. Targeting TNFR2 to overcome acquired adaptive resistance to immune checkpoint blockade
- Author
-
Lena Sophie Mayer, Robert J. Orlowski, Josephine Giles, Joseph L. Benci, Gavin Ellis, Guoping Deng, John Attanasio, Zeyu Chen, Bertram Bengsch, Omar Kahn, Sasikanth Manne, Ramin S. Herati, Shin Ngiow, Sangeeth M. George, Denise L. Faustman, Gary Gilliland, Rosemarie Mick, Wei Xu, Suzanne McGettigan, Xiaowei Xu, Ravi K. Amaravadi, Giorgos C. Karakousis, Lynn M. Schuchter, Tara C. Mitchell, James L. Riley, Alexander C. Huang, Andy Minn, Vesselin Tomov, and E. John Wherry
- Subjects
Immunology ,Immunology and Allergy - Abstract
Overcoming acquired adaptive immune resistance to anti-PD-1 therapy is imperative for enhancing the efficacy of immune checkpoint blockade (ICB) in solid tumors. Regulatory T cells (Tregs) play a prominent role in the suppressive tumor microenvironment (TME) and are major contributors to adaptive immune resistance. Tregs limit CD8+ T cell reinvigoration and are a promising target for combination therapy. While the clinical efficacy of anti-CTLA4 may be partially explained by restriction of Tregs, its co-administration with anti-PD1 causes significant toxicity. Thus, safer approaches to limit Treg activity are needed. To elucidate the dynamic changes in immuno-regulatory circuits within the TME during ICB, we performed deep immune profiling of peripheral blood and tumors from patients with advanced melanoma prior to (n=7) and after 1 cycle of anti-PD-1 therapy with pembrolizumab (n=9). Tregs were abundant in the TME and retained their immunosuppressive phenotype and functionality following anti-PD-1. Epigenetic, transcriptomic, and proteomic analysis of Tregs after ICB identified tumor necrosis factor receptor 2 (TNFR2) signaling as a possible driver of CD8+ T cell suppression. TNFR2 was preferentially expressed by Tregs in the TME (mean 18.03 %, SD +/− 10.13 %) relative to CD8+ T cells (mean 0.64 %, SD +/− 0.82 %) and peripheral Tregs (mean 3.16 %, SD +/− 3.21 %), suggesting it might be a safe and effective target for combination therapy. Indeed, dual blockade of TNFR2 and PD-1 led to potent CD8+ T cell expansion in two mouse tumor models, and restored sensitivity to ICB in a resistant murine model of melanoma. Our data suggest that anti-TNFR2 might synergize with current ICB by countering the development of adaptive immune resistance.
- Published
- 2020
18. PD1 and LAG3 converge to limit polyfunctionality and systemic immunity
- Author
-
Lawrence Peter Andrews, Sasikanth Manne, E. John Wherry, Creg Workman, and Dario Vignali
- Subjects
Immunology ,Immunology and Allergy - Abstract
Targeting PD1 has yielded clinical success across many tumor types, yet a significant proportion of patients remain unresponsive to treatment. Co-expression of PD1 and LAG3 inhibitory receptors (IRs) on CD8+ tumor-infiltrating T cells (TIL) is associated with an exhausted phenotype and dual blockade synergistically limits tumor growth, greater than targeting PD1 alone. The cellular and mechanistic basis for PD1/LAG3 synergy is unknown. We have generated conditional knockin mice that facilitates the CD8+ T cell-restricted deletion of Pdcd1 and/or Lag3 when crossed to E8ICre.GFP. These mice have also been crossed with a pMEL TCR transgenic mouse, congenically marked to allow intrinsic analysis of PD1 and/or LAG3-deficient CD8+ T cells, and controls, in a ‘quad’ co-adoptive transfer system. Pdcd1L/L and/or Lag3L/L-yfp E8ICre.GFP mice show reduced B16-F10 tumor growth with improved survival, compared to Lag3L/L-yfp E8ICre.GFP mice and controls. CD8+ TIL frequency is increased with loss of both IRs, as a result of enhanced proliferation. Although expression of TIM3, TIGIT and 2B4 IRs is maintained, CD8+ TIL isolated from Pdcd1L/LE8ICre.GFP and Pdcd1L/ Lag3L/L-yfp E8ICre.GFP mice show increased polyfunctionality, by IFNg, TNFa and GzmB release. This was confirmed to be largely driven by effector and chemoattractive secretions by analysis with a 28-plex single-cell cytokine response panel (Isoplexis). Overall, these data suggest that PD1 and LAG3 synergize to have a dominant effect on CD8+ TIL and limit antitumor immune effects, as intrinsic removal of both IRs results in reduced tumor growth that substantially impacts anti-tumor immunity. These results are encouraging for the development of co-targeting LAG3 and PD1 in the clinic.
- Published
- 2020
19. Developmental relationships of four exhausted CD8 T cell subsets reveals underlying transcriptional and epigenetic control mechanisms
- Author
-
Jean-Christophe Beltra, Sasikanth Manne, Mohamed S Abdel Hakeem, Makoto Kurachi, Josephine R Giles, Zeyu Chen, Valentina Casella, Shin Ngiow, Omar Khan, Yinghui Jane Huang, Patrick Yan, Kito Nzingha, Alexander C. Huang, and E. John Wherry
- Subjects
Immunology ,Immunology and Allergy - Abstract
Exhausted CD8 T cells (TEX) are essential during chronic viral infections and cancer. Two TEX subpopulations including a progenitor and a more terminally exhausted subset cooperate to maintain an active immune response during antigen persistence. However, non-overlapping delineations of these populations have suggested a more complex developmental biology. Here, using the LCMV mouse model of chronic viral infection, we identify four distinct TEX subsets based on Ly108 (Slamf6) and CD69 expression revealing a novel stepwise developmental framework. We reveal the transcriptional and epigenetic control mechanisms and associated biological changes underlying each TEX subset transition. Two TCF1+ progenitors were identified along with a novel TCF1-intermediate subset that re-engaged some aspects of effector biology. This subset depended on T-bet and was re-invigorated upon PD-L1 blockade. Ultimately, Tox coordinated loss of T-bet and differentiation into a fourth, terminally exhausted subset. These data define a new developmental hierarchy of TEX and reveal distinct biological properties with direct relevance to immunotherapy. Defining the control mechanisms of this TEX subset hierarchy provides novel opportunities to manipulate TEX biology for clinical goals.
- Published
- 2020
20. Developmental Relationships of Four Exhausted CD8+ T Cell Subsets Reveals Underlying Transcriptional and Epigenetic Landscape Control Mechanisms
- Author
-
Lynn M. Schuchter, Mohamed S. Abdel-Hakeem, E. John Wherry, Yinghui Jane Huang, Sasikanth Manne, Patrick Yan, Zeyu Chen, Kito Nzingha, Ravi K. Amaravadi, Valentina Casella, Shin Foong Ngiow, Wei Xu, Omar Khan, Jean-Christophe Beltra, Tara C. Mitchell, Makoto Kurachi, Giorgos C. Karakousis, Alexander C. Huang, Josephine R. Giles, and Xiaowei Xu
- Subjects
0301 basic medicine ,Effector ,medicine.medical_treatment ,Immunology ,Biology ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,Cancer immunotherapy ,030220 oncology & carcinogenesis ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Epigenetics ,Control (linguistics) ,Developmental biology ,CD8 ,Progenitor - Abstract
Summary CD8+ T cell exhaustion is a major barrier to current anti-cancer immunotherapies. Despite this, the developmental biology of exhausted CD8+ T cells (Tex) remains poorly defined, restraining improvement of strategies aimed at “re-invigorating” Tex cells. Here, we defined a four-cell-stage developmental framework for Tex cells. Two TCF1+ progenitor subsets were identified, one tissue restricted and quiescent and one more blood accessible, that gradually lost TCF1 as it divided and converted to a third intermediate Tex subset. This intermediate subset re-engaged some effector biology and increased upon PD-L1 blockade but ultimately converted into a fourth, terminally exhausted subset. By using transcriptional and epigenetic analyses, we identified the control mechanisms underlying subset transitions and defined a key interplay between TCF1, T-bet, and Tox in the process. These data reveal a four-stage developmental hierarchy for Tex cells and define the molecular, transcriptional, and epigenetic mechanisms that could provide opportunities to improve cancer immunotherapy.
- Published
- 2020
21. Mechanisms by which Obesity Dysregulates Immunometabolic State in Asthma
- Author
-
Peyton Conrey, Ramin S. Herati, Aaron Masino, Sasikanth Manne, Samir Sayed, De'Broski R. Herbert, Laura A. Vella, Hakon Hakonarson, Li-Yin Hung, Ting Qian, E. John Wherry, Kaitlin C. O'Boyle, Susan E. Coffin, Christopher F. Pastore, Joshua D. Rabinowitz, Frank D. Mentch, Bertram Bengsch, Scott E. Hensley, and Sarah E. Henrickson
- Subjects
business.industry ,Environmental health ,Immunology ,medicine ,Immunology and Allergy ,medicine.disease ,business ,Obesity ,Asthma - Published
- 2020
22. Epigenomic-Guided Mass Cytometry Profiling Reveals Disease-Specific Features of Exhausted CD8 T Cells
- Author
-
Omar Khan, Ramin S. Herati, Shaun O'Brien, Pier Federico Gherardini, Takuya Ohtani, Sasikanth Manne, Kyong-Mi Chang, Manu Setty, Dana Pe'er, Niels Bovenschen, Evan W. Newell, Steven M. Albelda, E. John Wherry, Bertram Bengsch, and Alexander C. Huang
- Subjects
Epigenomics ,Proteomics ,0301 basic medicine ,mass cytometry ,Lung Neoplasms ,medicine.medical_treatment ,Immunology ,HIV Infections ,Computational biology ,cancer immunology ,CD8 T cell ,chronic infection ,CyTOF ,HIV ,immune checkpoint ,lung cancer ,systems immunology ,T cell exhaustion ,CD8-Positive T-Lymphocytes ,Biology ,complex mixtures ,Article ,Transcriptome ,03 medical and health sciences ,medicine ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Mass cytometry ,Cancer immunology ,Settore BIO/11 ,Gene Expression Profiling ,virus diseases ,Immunotherapy ,Flow Cytometry ,Gene expression profiling ,030104 developmental biology ,Infectious Diseases ,human activities ,Transcription Factors - Abstract
Exhausted CD8 T (Tex) cells are immunotherapy targets in chronic infection and cancer, but a comprehensive assessment of Tex cell diversity in human disease is lacking. Here, we developed a transcriptomic- and epigenetic-guided mass cytometry approach to define core exhaustion-specific genes and disease-induced changes in Tex cells in HIV and human cancer. Single-cell proteomic profiling identified 9 distinct Tex cell clusters using phenotypic, functional, transcription factor, and inhibitory receptor co-expression patterns. An exhaustion severity metric was developed and integrated with high-dimensional phenotypes to define Tex cell clusters that were present in healthy subjects, common across chronic infection and cancer or enriched in either disease, linked to disease severity, and changed with HIV therapy. Combinatorial patterns of immunotherapy targets on different Tex cell clusters were also defined. This approach and associated datasets present a resource for investigating human Tex cell biology, with implications for immune monitoring and immunomodulation in chronic infections, autoimmunity, and cancer. Exhausted T (Tex) cells have poor function in chronic infections and cancer but can be therapeutically re-invigorated. Bengsch et al. use genes modified epigenetically during exhaustion and high-dimensional CyTOF profiling to define Tex cell heterogeneity in humans with HIV or lung cancer and link Tex cell features to disease progression and response to immunotherapy.
- Published
- 2018
23. Differentiation and protective capacity of virus-specific CD8+ T cells suggest murine norovirus persistence in an immune-privileged enteric niche
- Author
-
Laurence C. Eisenlohr, Gabriela L. Cosma, Ralitza Tacheva, Herbert W. Virgin, Vesselin T. Tomov, Ajinkya Pattekar, Chi Wai Lau, Timothy J. Nice, Olesya Palko, E. John Wherry, Yuhang Sun, Bertram Bengsch, and Sasikanth Manne
- Subjects
0301 basic medicine ,T cell ,Immunology ,ved/biology.organism_classification_rank.species ,CD8-Positive T-Lymphocytes ,Biology ,Adaptive Immunity ,Article ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Viral shedding ,Caliciviridae Infections ,Oligonucleotide Array Sequence Analysis ,Immune Evasion ,ved/biology ,Gene Expression Profiling ,Norovirus ,Cell Differentiation ,Epithelial Cells ,Virology ,Immunity, Innate ,Gastroenteritis ,Mice, Inbred C57BL ,Chronic infection ,Gene Ontology ,HEK293 Cells ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Cellular Microenvironment ,Viral replication ,Host-Pathogen Interactions ,Immunologic Memory ,CD8 ,030215 immunology ,Murine norovirus - Abstract
Noroviruses can establish chronic infections with active viral shedding in healthy humans but whether persistence is associated with adaptive immune dysfunction is unknown. We used genetically engineered strains of mouse norovirus (MNV) to investigate CD8+ T cell differentiation during chronic infection. We found that chronic infection drove MNV-specific tissue-resident memory (Trm) CD8+ T cells to a differentiation state resembling inflationary effector responses against latent cytomegalovirus with only limited evidence of exhaustion. These MNV-specific Trm cells remained highly functional yet appeared ignorant of ongoing viral replication. Pre-existing MNV-specific Trm cells provided partial protection against chronic infection but largely ceased to detect virus within 72 hours of challenge, demonstrating rapid sequestration of viral replication away from T cells. Our studies revealed a strategy of immune evasion by MNV via the induction of a CD8+ T cell program normally reserved for latent pathogens and persistence in an immune-privileged enteric niche.
- Published
- 2017
24. OBESITY DYSREGULATES IMMUNOMETABOLIC STATUS IN ASTHMA AND IMPACTS VACCINE RESPONSES
- Author
-
Susan E. Coffin, E. John Wherry, Laura A. Vella, Josh Rabinowitz, Scott E. Hensley, Sasikanth Manne, Bertram Bengsch, Sarah E. Henrickson, Kaitlin C. O'Boyle, and Ramin S. Herati
- Subjects
business.industry ,Environmental health ,Immunology ,Immunology and Allergy ,Medicine ,business ,medicine.disease ,Obesity ,Asthma - Published
- 2019
25. Modulation of TFH-like cells and anti-tumor activity of immune checkpoint blockade
- Author
-
Roberta Zappasodi, Sadna Budhu, Matthew D. Hellmann, Michael A. Postow, Yasin Senbabaoglu, Sasikanth Manne, Billel Gasmi, Cailian Liu, Hong Zhong, Yanyun Li, Alexander C. Huang, Daniel Hirschhorn-Cymerman, Katherine S. Panageas, E. John Wherry, Taha Merghoub, and Jedd D. Wolchok
- Subjects
Immunology ,Immunology and Allergy - Abstract
A significant proportion of cancer patients do not respond to immune checkpoint blockade therapy. To deepen our understanding of the mechanisms of resistance to immunotherapy, we studied a population of CD4+Foxp3− T cells expressing PD-1 (4PD1hi), which we found to be up-regulated in B16-melanoma bearing mice after CTLA-4 blockade in association with limited response to treatment. We observed that 4PD1hi accumulate intratumorally as a function of tumor burden in untreated tumor-bearing hosts. Interestingly, CTLA-4 blockade promotes intratumoral and peripheral 4PD1hiincreases in a dose-dependent manner, while combination with PD-1 blockade mitigates this effect in both B16-bearing mice and melanoma patients and significantly improves anti-tumor activity. In addition, we found that persistence of high levels of 4PD1hi after PD-1 blockade correlates with poor prognosis in melanoma patients. Mechanistically, we show that mouse and human circulating and intra-tumor 4PD1hi inhibit T-cell functions in a PD-1/PD-L1 dependent fashion. In addition, we found that mouse and human 4PD1hi resemble follicular-helper-T-cell(TFH)-like cells and that CTLA-4 blockade activity is improved in TFH deficient mice. These findings broaden our understanding of the mechanisms that limit anti-tumor immunity, providing an additional explanation for the incremental activity of combined CTLA-4 and PD-1 blockade. Our study also defines 4PD1hi as a new prognostic and pharmacodynamic biomarker for the design of optimal checkpoint blockade combination schedules and dosage.
- Published
- 2018
26. Chronic Allergen Stimulation Yields T cell Dysfunction in Obese Asthmatics
- Author
-
E. John Wherry, Joshua R. Rabinowitz, Laura A. Vella, Ramin S. Herati, Susan E. Coffin, Sasikanth Manne, Kaitlin C. O'Boyle, Bertram Bengsch, Sarah E. Henrickson, and Scott E. Hensley
- Subjects
T-cell dysfunction ,Allergen ,business.industry ,Immunology ,Immunology and Allergy ,Medicine ,Stimulation ,business ,medicine.disease_cause - Published
- 2018
27. Metabolic and Deep Immune Profiling Reveal Coordinate Effects on Immune Function in Asthma and Obesity
- Author
-
Scott E. Hensley, Laura A. Vella, Joshua D. Rabinowitz, Sasikanth Manne, Sarah E. Henrickson, Bertram Bengsch, Ramin S. Herati, Susan E. Coffin, and E. John Wherry
- Subjects
Immune profiling ,Immune system ,business.industry ,Immunology ,medicine ,Immunology and Allergy ,medicine.disease ,business ,Obesity ,Asthma - Published
- 2017
28. MiR-150 negatively regulates CD8+ T cell memory formation
- Author
-
Zeyu Chen, Erietta Stelekati, Makoto Kurachi, Sixiang Yu, Sasikanth Manne, and E. John Wherry
- Subjects
Immunology ,Immunology and Allergy - Abstract
MicroRNAs play an important role in CD8+ T cell differentiation and anti-viral immune responses. However, how microRNAs regulate CD8+ T cell memory formation remains poorly defined. Here, by microRNA profiling of CD8+ T cell during acute or chronic LCMV infection, we identified microRNAs that are specifically enriched in memory CD8+ T cells comparing to naïve, effector or exhausted CD8+ T cells. MiR-150, a microRNA that has been shown to be important in multiple biological processes, was identified as a potential memory CD8+ T cell biased microRNA. Functional interrogation of the role of miR-150 demonstrated that absence of miR-150 in CD8+ T cells enhanced memory CD8+ T cell differentiation, by both increasing KLRG-CD127+ memory precursors population during effector phase and enhancing CD127+CXCR3+ central memory phenotype long-term. Overexpression of miR-150 in CD8+ T cells, in contrast, negatively regulated CD8+ T cell memory formation. Furthermore, we found that miR-150 KO CD8+ T cells had higher expression of c-Myb comparing to miR-150 WT CD8+ T cells, a result observed using both in vitro CD8+ T cell differentiation and following infection with LCMV-Armstrong in vivo. Gene expression analysis implicated a relationship between the level of c-Myb expression and CD8+ T cell memory formation through potential anti-apoptotic molecules such as bcl-2. Thus, our studies identify a key role for miR-150 in CD8+ T cell memory and implicate a mechanism of CD8+ T cell memory effect by this microRNA through regulation of c-Myb.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.