53 results on '"Schenkel JM"'
Search Results
2. In vivo antibody labeling route and fluorophore dictate labeling efficiency, sensitivity, and longevity.
- Author
-
Hagan NB, Inaku C, Kunder N, White T, Iraguha T, Meyer A, Pauken KE, and Schenkel JM
- Abstract
Leukocytes migrate through the blood and extravasate into organs to surveil the host for infection or cancer. Recently, we demonstrated that intravenous (IV) anti-CD45.2 antibody labeling allowed for precise tracking of leukocyte migration. However, the narrow labeling window can make this approach challenging for tracking rare migration events. Here, we show that altering antibody administration route and fluorophore can significantly extend the antibody active labeling time. We found that while both IV and intraperitoneal (IP) anti-CD45.2 antibody labeled circulating leukocytes after injection, they had different kinetic properties that impacted labeling time and intensity. Quantification of circulating antibody revealed that while unbound IV anti-CD45.2 antibody rapidly decreased, unbound IP anti-CD45.2 antibody increased over one hour. Using in vitro and in vivo serial dilution assays, we found that Alexa Fluor 647 (AF647) and Brilliant Blue 700 (BB700) dyes had the greatest labeling sensitivity compared to other fluorophores. However, IP antibody injection with anti-CD45.2 BB700, but not AF647, resulted in continuous blood leukocyte labeling for over 6 hours. Finally, we leveraged IP anti-CD45.2 BB700 antibody to track slower migrating leukocytes into tumors. We found that IP anti-CD45.2 antibody injection allowed for the identification of ~seven times as many tumor-specific CD8
+ T cells that had recently migrated from blood into tumors. Our results demonstrate how different injection routes and fluorophores affect anti-CD45.2 antibody leukocyte labeling and highlight the utility of this approach for defining leukocyte migration in the context of homeostasis and cancer., Competing Interests: Competing Interests The authors declare that they have no competing interests.- Published
- 2024
- Full Text
- View/download PDF
3. Longitudinal Intravascular Antibody Labeling Identified Regulatory T Cell Recruitment as a Therapeutic Target in a Mouse Model of Lung Cancer.
- Author
-
Shanahan SL, Kunder N, Inaku C, Hagan NB, Gibbons G, Mathey-Andrews N, Anandappa G, Soares S, Pauken KE, Jacks T, and Schenkel JM
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Tumor Microenvironment immunology, Adenocarcinoma immunology, Cell Line, Tumor, T-Lymphocytes, Regulatory immunology, Lung Neoplasms immunology, Lung Neoplasms pathology, Disease Models, Animal, Cell Movement immunology
- Abstract
Anticancer immunity is predicated on leukocyte migration into tumors. Once recruited, leukocytes undergo substantial reprogramming to adapt to the tumor microenvironment. A major challenge in the field is distinguishing recently recruited from resident leukocytes in tumors. In this study, we developed an intravascular Ab technique to label circulating mouse leukocytes before they migrate to tissues, providing unprecedented insight into the kinetics of recruitment. This approach unveiled the substantial role of leukocyte migration in tumor progression using a preclinical mouse model of lung adenocarcinoma. Regulatory T cells (Tregs), critical mediators of immunosuppression, were continuously and rapidly recruited into tumors throughout cancer progression. Moreover, leukocyte trafficking depended on the integrins CD11a/CD49d, and CD11a/CD49d blockade led to significant tumor burden reduction in mice. Importantly, preventing circulating Treg recruitment through depletion or sequestration in lymph nodes was sufficient to decrease tumor burden, indicating that Treg migration was crucial for suppressing antitumor immunity. These findings underscore the dynamic nature of the immune compartment within mouse lung tumors and demonstrate the relevance of a temporal map of leukocyte recruitment into tumors, thereby advancing our understanding of leukocyte migration in the context of tumor development., (Copyright © 2024 by The American Association of Immunologists, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
4. Localization, tissue biology and T cell state - implications for cancer immunotherapy.
- Author
-
Schenkel JM and Pauken KE
- Subjects
- Humans, Immunologic Memory, Immunotherapy, Biology, Tumor Microenvironment, CD8-Positive T-Lymphocytes, Neoplasms
- Abstract
Tissue localization is a critical determinant of T cell immunity. CD8
+ T cells are contact-dependent killers, which requires them to physically be within the tissue of interest to kill peptide-MHC class I-bearing target cells. Following their migration and extravasation into tissues, T cells receive many extrinsic cues from the local microenvironment, and these signals shape T cell differentiation, fate and function. Because major organ systems are variable in their functions and compositions, they apply disparate pressures on T cells to adapt to the local microenvironment. Additional complexity arises in the context of malignant lesions (either primary or metastatic), and this has made understanding the factors that dictate T cell function and longevity in tumours challenging. Moreover, T cell differentiation state influences how cues from the microenvironment are interpreted by tissue-infiltrating T cells, highlighting the importance of T cell state in the context of tissue biology. Here, we review the intertwined nature of T cell differentiation state, location, survival and function, and explain how dysfunctional T cell populations can adopt features of tissue-resident memory T cells to persist in tumours. Finally, we discuss how these factors have shaped responses to cancer immunotherapy., (© 2023. Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
5. Tissue-resident memory T cells trigger rapid exudation and local antibody accumulation.
- Author
-
Rosato PC, Lotfi-Emran S, Joag V, Wijeyesinghe S, Quarnstrom CF, Degefu HN, Nedellec R, Schenkel JM, Beura LK, Hangartner L, Burton DR, and Masopust D
- Subjects
- Mice, Animals, Female, Memory T Cells, Immunoglobulins, Immunologic Memory, CD8-Positive T-Lymphocytes, Stomatitis
- Abstract
Adaptive immunity is didactically partitioned into humoral and cell-mediated effector mechanisms, which may imply that each arm is separate and does not function together. Here, we report that the activation of CD8+ resident memory T cells (T
RM ) in nonlymphoid tissues triggers vascular permeability, which facilitates rapid distribution of serum antibodies into local tissues. TRM reactivation was associated with transcriptional upregulation of antiviral signaling pathways as well as Fc receptors and components of the complement cascade. Effects were local, but evidence is presented that TRM in brain and reproductive mucosa are both competent to induce rapid antibody exudation. TRM reactivation in the mouse female genital tract increased local concentrations of virus-specific neutralizing antibodies, including anti-vesicular stomatitis virus, and passively transferred anti-HIV antibodies. We showed that this response was sufficient to increase the efficacy of ex vivo vesicular stomatitis virus neutralization. These results indicate that CD8+ TRM antigen recognition can enhance local humoral immunity., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
6. Modeling diverse genetic subtypes of lung adenocarcinoma with a next-generation alveolar type 2 organoid platform.
- Author
-
Naranjo S, Cabana CM, LaFave LM, Romero R, Shanahan SL, Bhutkar A, Westcott PMK, Schenkel JM, Ghosh A, Liao LZ, Del Priore I, Yang D, and Jacks T
- Subjects
- Animals, Humans, Mice, Organoids, Proto-Oncogene Proteins B-raf metabolism, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Receptor Protein-Tyrosine Kinases metabolism, Adenocarcinoma of Lung genetics, Adenocarcinoma of Lung metabolism, Adenocarcinoma of Lung pathology, Lung Neoplasms metabolism
- Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Lung adenocarcinoma (LUAD), the most common histological subtype, accounts for 40% of all cases. While existing genetically engineered mouse models (GEMMs) recapitulate the histological progression and transcriptional evolution of human LUAD, they are time-consuming and technically demanding. In contrast, cell line transplant models are fast and flexible, but these models fail to capture the full spectrum of disease progression. Organoid technologies provide a means to create next-generation cancer models that integrate the most advantageous features of autochthonous and transplant-based systems. However, robust and faithful LUAD organoid platforms are currently lacking. Here, we describe optimized conditions to continuously expand murine alveolar type 2 (AT2) cells, a prominent cell of origin for LUAD, in organoid culture. These organoids display canonical features of AT2 cells, including marker gene expression, the presence of lamellar bodies, and an ability to differentiate into the AT1 lineage. We used this system to develop flexible and versatile immunocompetent organoid-based models of KRAS , BRAF , and ALK mutant LUAD. Notably, organoid-based tumors display extensive burden and complete penetrance and are histopathologically indistinguishable from their autochthonous counterparts. Altogether, this organoid platform is a powerful, versatile new model system to study LUAD., (© 2022 Naranjo et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2022
- Full Text
- View/download PDF
7. Single-nucleus and spatial transcriptome profiling of pancreatic cancer identifies multicellular dynamics associated with neoadjuvant treatment.
- Author
-
Hwang WL, Jagadeesh KA, Guo JA, Hoffman HI, Yadollahpour P, Reeves JW, Mohan R, Drokhlyansky E, Van Wittenberghe N, Ashenberg O, Farhi SL, Schapiro D, Divakar P, Miller E, Zollinger DR, Eng G, Schenkel JM, Su J, Shiau C, Yu P, Freed-Pastor WA, Abbondanza D, Mehta A, Gould J, Lambden C, Porter CBM, Tsankov A, Dionne D, Waldman J, Cuoco MS, Nguyen L, Delorey T, Phillips D, Barth JL, Kem M, Rodrigues C, Ciprani D, Roldan J, Zelga P, Jorgji V, Chen JH, Ely Z, Zhao D, Fuhrman K, Fropf R, Beechem JM, Loeffler JS, Ryan DP, Weekes CD, Ferrone CR, Qadan M, Aryee MJ, Jain RK, Neuberg DS, Wo JY, Hong TS, Xavier R, Aguirre AJ, Rozenblatt-Rosen O, Mino-Kenudson M, Castillo CF, Liss AS, Ting DT, Jacks T, and Regev A
- Subjects
- Biomarkers, Tumor genetics, Gene Expression Profiling, Humans, Neoadjuvant Therapy, Prognosis, Transcriptome genetics, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology, Carcinoma, Pancreatic Ductal therapy, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal and treatment-refractory cancer. Molecular stratification in pancreatic cancer remains rudimentary and does not yet inform clinical management or therapeutic development. Here, we construct a high-resolution molecular landscape of the cellular subtypes and spatial communities that compose PDAC using single-nucleus RNA sequencing and whole-transcriptome digital spatial profiling (DSP) of 43 primary PDAC tumor specimens that either received neoadjuvant therapy or were treatment naive. We uncovered recurrent expression programs across malignant cells and fibroblasts, including a newly identified neural-like progenitor malignant cell program that was enriched after chemotherapy and radiotherapy and associated with poor prognosis in independent cohorts. Integrating spatial and cellular profiles revealed three multicellular communities with distinct contributions from malignant, fibroblast and immune subtypes: classical, squamoid-basaloid and treatment enriched. Our refined molecular and cellular taxonomy can provide a framework for stratification in clinical trials and serve as a roadmap for therapeutic targeting of specific cellular phenotypes and multicellular interactions., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
8. Deciphering the immunopeptidome in vivo reveals new tumour antigens.
- Author
-
Jaeger AM, Stopfer LE, Ahn R, Sanders EA, Sandel DA, Freed-Pastor WA, Rideout WM 3rd, Naranjo S, Fessenden T, Nguyen KB, Winter PS, Kohn RE, Westcott PMK, Schenkel JM, Shanahan SL, Shalek AK, Spranger S, White FM, and Jacks T
- Subjects
- Alveolar Epithelial Cells immunology, Animals, Antigen Presentation, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, Carcinoma, Pancreatic Ductal chemistry, Carcinoma, Pancreatic Ductal immunology, Histocompatibility Antigens Class I genetics, Histocompatibility Antigens Class I immunology, Lung Neoplasms chemistry, Lung Neoplasms immunology, Mice, Pancreatic Neoplasms chemistry, Pancreatic Neoplasms immunology, RNA, Messenger, Antigens, Neoplasm analysis, Antigens, Neoplasm chemistry, Antigens, Neoplasm immunology, Peptides analysis, Peptides chemistry, Peptides immunology, Proteomics
- Abstract
Immunosurveillance of cancer requires the presentation of peptide antigens on major histocompatibility complex class I (MHC-I) molecules
1-5 . Current approaches to profiling of MHC-I-associated peptides, collectively known as the immunopeptidome, are limited to in vitro investigation or bulk tumour lysates, which limits our understanding of cancer-specific patterns of antigen presentation in vivo6 . To overcome these limitations, we engineered an inducible affinity tag into the mouse MHC-I gene (H2-K1) and targeted this allele to the KrasLSL-G12D/+ Trp53fl/fl mouse model (KP/Kb Strep)7 . This approach enabled us to precisely isolate MHC-I peptides from autochthonous pancreatic ductal adenocarcinoma and from lung adenocarcinoma (LUAD) in vivo. In addition, we profiled the LUAD immunopeptidome from the alveolar type 2 cell of origin up to late-stage disease. Differential peptide presentation in LUAD was not predictable by mRNA expression or translation efficiency and is probably driven by post-translational mechanisms. Vaccination with peptides presented by LUAD in vivo induced CD8+ T cell responses in naive mice and tumour-bearing mice. Many peptides specific to LUAD, including immunogenic peptides, exhibited minimal expression of the cognate mRNA, which prompts the reconsideration of antigen prediction pipelines that triage peptides according to transcript abundance8 . Beyond cancer, the Kb Strep allele is compatible with other Cre-driver lines to explore antigen presentation in vivo in the pursuit of understanding basic immunology, infectious disease and autoimmunity., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
9. Therapeutic avenues for cancer neuroscience: translational frontiers and clinical opportunities.
- Author
-
Shi DD, Guo JA, Hoffman HI, Su J, Mino-Kenudson M, Barth JL, Schenkel JM, Loeffler JS, Shih HA, Hong TS, Wo JY, Aguirre AJ, Jacks T, Zheng L, Wen PY, Wang TC, and Hwang WL
- Subjects
- Cancer Pain drug therapy, Clinical Trials as Topic, Drug Resistance, Neoplasm, Humans, Immune Checkpoint Inhibitors therapeutic use, Neoplasms etiology, Neoplasms immunology, Neoplasms pathology, Nervous System Physiological Phenomena drug effects, Tumor Microenvironment, Neoplasms drug therapy, Neurosciences
- Abstract
With increasing attention on the essential roles of the tumour microenvironment in recent years, the nervous system has emerged as a novel and crucial facilitator of cancer growth. In this Review, we describe the foundational, translational, and clinical advances illustrating how nerves contribute to tumour proliferation, stress adaptation, immunomodulation, metastasis, electrical hyperactivity and seizures, and neuropathic pain. Collectively, this expanding knowledge base reveals multiple therapeutic avenues for cancer neuroscience that warrant further exploration in clinical studies. We discuss the available clinical data, including ongoing trials investigating novel agents targeting the tumour-nerve axis, and the therapeutic potential for repurposing existing neuroactive drugs as an anti-cancer approach, particularly in combination with established treatment regimens. Lastly, we discuss the clinical challenges of these treatment strategies and highlight unanswered questions and future directions in the burgeoning field of cancer neuroscience., Competing Interests: Declaration of interests MM-K reports personal fees from H3 Biomedicine and AstraZeneca; grants from Novartis; and royalties from Elsevier, outside the submitted work. TSH reports personal fees from Synthetic Biologics, Novocure, and Merck; equity in PanTher Therapeutics; and grants from Taiho, AstraZeneca, Bristol Myers Squibb, Tesaro, Ipsen, and Puma, outside the submitted work. AJA reports personal fees from Merck, Arrakis Therapeutics, and Oncorus; grants from Mirati Therapeutics, Deerfield, Novo Ventures, National Cancer Institute, Lustgarten Foundation, Pancreatic Cancer Action Network, Doris Duke Charitable Foundation, and Dana-Farber Cancer Institute Hale Center for Pancreatic Cancer Research; and grants and personal fees from Syros Pharmaceuticals, outside the submitted work. TJ reports grants from Lustgarten Foundation; stock in Amgen and Thermo Fisher Scientific; and membership of the Board of Directors of Amgen and Thermo Fisher Scientific, outside the submitted work. LZ reports grants from Merck, during the conduct of the study; and grants from iTeos, Bristol Meyers Squibb, Merck, AstraZeneca, Amgen, NovaRock, Inxmed, and Halozyme; personal fees from Biosion, NovaRock, Akrevia/ Xilio, Datarevive, QED, Natera, Ambrx, Snow Lake Capital, and Tempus; personal fees from Alphamab and Mingruizhiyao; and shares in Alphamab and Mingruizhiyao, outside the submitted work. PYW reports research support from Agios, AstraZeneca/Medimmune, Beigene, Celgene, Eli Lily, Genentech/Roche, Kazia, MediciNova, Merck, Novartis, Nuvation Bio, Oncoceutics, Vascular Biogenics, and VBI Vaccines; and membership on the advisory board of Agios, AstraZeneca, Bayer, Boston Pharmaceuticals, CNS Pharmaceuticals, Elevate Bio Immunomic Therapeutics, Imvax, Karyopharm, Merck, Novartis, Nuvation Bio, Vascular Biogenics, VBI Vaccines, and Voyager, outside the submitted work. All other authors declare no competing interests., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
10. Mental health, art, and deinstitutionalization: an aesthetic-poetic-theatrical account of the city's occupation.
- Author
-
Schenkel JM, Silva GWDS, Amorin AKMA, Miranda FAN, Carvalho JBL, Ribeiro SEA, Almeida ACP, and Silva MM
- Subjects
- Brazil, Deinstitutionalization, Esthetics, Humans, Occupations, Mental Health, Mental Health Services
- Abstract
This study aimed to report the experience of an aesthetic, poetic, and theatrical production of the city's occupation from a device of the Psychosocial Care Network to offer space for sociability, production, and cultural intervention. This is an account of an experience from the Social and Cultural Center (CECCO) in Natal, Rio Grande do Norte, Brazil, within madness and mental health deinstitutionalization. The intervention "The Little Prince occupies the Ribeira" was inspired by the work of author Saint-Exupéry. The artistic and creative acts reported occurred in December 2019. We experienced in this intervention the reach of an aesthetic clinic that, when opened to the street and art, expanded and weaved in the territory, instrumentalized by theater, dance, poetry, percussion, crafts, and city's occupation. The movement led the community to "step down from the stage" to the streets and is connected with the twist of the asylum model and the production of the affection clinic that we seek to sustain in the daily service.
- Published
- 2022
- Full Text
- View/download PDF
11. Conventional type I dendritic cells maintain a reservoir of proliferative tumor-antigen specific TCF-1 + CD8 + T cells in tumor-draining lymph nodes.
- Author
-
Schenkel JM, Herbst RH, Canner D, Li A, Hillman M, Shanahan SL, Gibbons G, Smith OC, Kim JY, Westcott P, Hwang WL, Freed-Pastor WA, Eng G, Cuoco MS, Rogers P, Park JK, Burger ML, Rozenblatt-Rosen O, Cong L, Pauken KE, Regev A, and Jacks T
- Subjects
- Animals, Mice, T-Lymphocyte Subsets immunology, Adenocarcinoma of Lung immunology, CD8-Positive T-Lymphocytes immunology, Dendritic Cells immunology, Lung Neoplasms immunology, Lymph Nodes immunology, T Cell Transcription Factor 1 immunology
- Abstract
In tumors, a subset of CD8
+ T cells expressing the transcription factor TCF-1 drives the response to immune checkpoint blockade. We examined the mechanisms that maintain these cells in an autochthonous model of lung adenocarcinoma. Longitudinal sampling and single-cell sequencing of tumor-antigen specific TCF-1+ CD8+ T cells revealed that while intratumoral TCF-1+ CD8+ T cells acquired dysfunctional features and decreased in number as tumors progressed, TCF-1+ CD8+ T cell frequency in the tumor draining LN (dLN) remained stable. Two discrete intratumoral TCF-1+ CD8+ T cell subsets developed over time-a proliferative SlamF6+ subset and a non-cycling SlamF6- subset. Blocking dLN egress decreased the frequency of intratumoral SlamF6+ TCF-1+ CD8+ T cells. Conventional type I dendritic cell (cDC1) in dLN decreased in number with tumor progression, and Flt3L+anti-CD40 treatment recovered SlamF6+ T cell frequencies and decreased tumor burden. Thus, cDC1s in tumor dLN maintain a reservoir of TCF-1+ CD8+ T cells and their decrease contributes to failed anti-tumor immunity., Competing Interests: Declaration of interests T.J. is a member of the Board of Directors of Amgen and Thermo Fisher Scientific. He is also a co-founder of Dragonfly Therapeutics and T2 Biosystems. T.J. serves on the Scientific Advisory Board of Dragonfly Therapeutics, SQZ Biotech, and Skyhawk Therapeutics. He is the President of Break Through Cancer. None of these affiliations represent a conflict of interest with respect to the design or execution of this study or interpretation of data presented in this manuscript. T.J. laboratory currently also receives funding from the Johnson & Johnson Lung Cancer Initiative and The Lustgarten Foundation for Pancreatic Cancer Research, but this funding did not support the research described in this manuscript. A.R. is a co-founder and equity holder of Celsius Therapeutics, an equity holder in Immunitas, and was an SAB member of ThermoFisher Scientific, Syros Pharmaceuticals, Neogene Therapeutics and Asimov until July 31, 2020. From August 1, 2020, A.R. is an employee of Genentech., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
12. The CD155/TIGIT axis promotes and maintains immune evasion in neoantigen-expressing pancreatic cancer.
- Author
-
Freed-Pastor WA, Lambert LJ, Ely ZA, Pattada NB, Bhutkar A, Eng G, Mercer KL, Garcia AP, Lin L, Rideout WM 3rd, Hwang WL, Schenkel JM, Jaeger AM, Bronson RT, Westcott PMK, Hether TD, Divakar P, Reeves JW, Deshpande V, Delorey T, Phillips D, Yilmaz OH, Regev A, and Jacks T
- Subjects
- Animals, Humans, Mice, Pancreatic Neoplasms, Immune Evasion immunology, Immunotherapy methods, Lymphocytes, Tumor-Infiltrating metabolism, Pancreatic Neoplasms immunology, Receptors, Virus immunology
- Abstract
The CD155/TIGIT axis can be co-opted during immune evasion in chronic viral infections and cancer. Pancreatic adenocarcinoma (PDAC) is a highly lethal malignancy, and immune-based strategies to combat this disease have been largely unsuccessful to date. We corroborate prior reports that a substantial portion of PDAC harbors predicted high-affinity MHC class I-restricted neoepitopes and extend these findings to advanced/metastatic disease. Using multiple preclinical models of neoantigen-expressing PDAC, we demonstrate that intratumoral neoantigen-specific CD8
+ T cells adopt multiple states of dysfunction, resembling those in tumor-infiltrating lymphocytes of PDAC patients. Mechanistically, genetic and/or pharmacologic modulation of the CD155/TIGIT axis was sufficient to promote immune evasion in autochthonous neoantigen-expressing PDAC. Finally, we demonstrate that the CD155/TIGIT axis is critical in maintaining immune evasion in PDAC and uncover a combination immunotherapy (TIGIT/PD-1 co-blockade plus CD40 agonism) that elicits profound anti-tumor responses in preclinical models, now poised for clinical evaluation., Competing Interests: Declaration of interests T.J. is a member of the Board of Directors of Amgen and Thermo Fisher Scientific, and a co-Founder of Dragonfly Therapeutics and T2 Biosystems. T.J. serves on the Scientific Advisory Board of Dragonfly Therapeutics, SQZ Biotech, and Skyhawk Therapeutics. T.J. is also President of Break Through Cancer. His laboratory currently receives funding from Johnson & Johnson and The Lustgarten Foundation; funds from the Lustgarten Foundation supported the research described in this manuscript. A.R. is a founder and equity holder of Celsius Therapeutics, an equity holder in Immunitas Therapeutics and until August 31, 2020, was an SAB member of Syros Pharmaceuticals, Neogene Therapeutics, Asimov and Thermo Fisher Scientific. From August 1, 2020, A.R. is an employee of Genentech, a member of the Roche Group. A.R. and Regev lab members' work was conducted at the Broad Institute, unrelated to these later affiliations. T.D.H., P.D., and J.W.R. are employees and stockholders at NanoString Technologies, Inc. None of these affiliations represent a conflict of interest with respect to the design or execution of this study or interpretation of data presented in this manuscript., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
13. Low neoantigen expression and poor T-cell priming underlie early immune escape in colorectal cancer.
- Author
-
Westcott PMK, Sacks NJ, Schenkel JM, Ely ZA, Smith O, Hauck H, Jaeger AM, Zhang D, Backlund CM, Beytagh MC, Patten JJ, Elbashir R, Eng G, Irvine DJ, Yilmaz OH, and Jacks T
- Subjects
- Animals, Antigens, Neoplasm genetics, Humans, Immunotherapy, Mice, Microsatellite Instability, Colorectal Neoplasms genetics, T-Lymphocytes
- Abstract
Immune evasion is a hallmark of cancer, and therapies that restore immune surveillance have proven highly effective in cancers with high tumor mutation burden (TMB) (e.g., those with microsatellite instability (MSI)). Whether low TMB cancers, which are largely refractory to immunotherapy, harbor potentially immunogenic neoantigens remains unclear. Here, we show that tumors from all patients with microsatellite stable (MSS) colorectal cancer (CRC) express clonal predicted neoantigens despite low TMB. Unexpectedly, these neoantigens are broadly expressed at lower levels compared to those in MSI CRC. Using a versatile platform for modulating neoantigen expression in CRC organoids and transplantation into the distal colon of mice, we show that low expression precludes productive cross priming and drives immediate T cell dysfunction. Strikingly, experimental or therapeutic rescue of priming rendered T cells capable of controlling tumors with low neoantigen expression. These findings underscore a critical role of neoantigen expression level in immune evasion and therapy response.
- Published
- 2021
- Full Text
- View/download PDF
14. Antigen dominance hierarchies shape TCF1 + progenitor CD8 T cell phenotypes in tumors.
- Author
-
Burger ML, Cruz AM, Crossland GE, Gaglia G, Ritch CC, Blatt SE, Bhutkar A, Canner D, Kienka T, Tavana SZ, Barandiaran AL, Garmilla A, Schenkel JM, Hillman M, de Los Rios Kobara I, Li A, Jaeger AM, Hwang WL, Westcott PMK, Manos MP, Holovatska MM, Hodi FS, Regev A, Santagata S, and Jacks T
- Subjects
- Amino Acid Sequence, Animals, CTLA-4 Antigen metabolism, Epitopes, Female, Humans, Immune Checkpoint Inhibitors pharmacology, Lung Neoplasms pathology, Mice, Peptides chemistry, Phenotype, Programmed Cell Death 1 Receptor metabolism, RNA-Seq, Receptors, Antigen, T-Cell metabolism, Receptors, CCR6 metabolism, Single-Cell Analysis, Vaccination, Adenocarcinoma of Lung immunology, Antigens, Neoplasm immunology, CD8-Positive T-Lymphocytes immunology, Hepatocyte Nuclear Factor 1-alpha metabolism, Lung Neoplasms immunology, Stem Cells immunology
- Abstract
CD8 T cell responses against different tumor neoantigens occur simultaneously, yet little is known about the interplay between responses and its impact on T cell function and tumor control. In mouse lung adenocarcinoma, we found that immunodominance is established in tumors, wherein CD8 T cell expansion is predominantly driven by the antigen that most stably binds MHC. T cells responding to subdominant antigens were enriched for a TCF1
+ progenitor phenotype correlated with response to immune checkpoint blockade (ICB) therapy. However, the subdominant T cell response did not preferentially benefit from ICB due to a dysfunctional subset of TCF1+ cells marked by CCR6 and Tc17 differentiation. Analysis of human samples and sequencing datasets revealed that CCR6+ TCF1+ cells exist across human cancers and are not correlated with ICB response. Vaccination eliminated CCR6+ TCF1+ cells and dramatically improved the subdominant response, highlighting a strategy to optimally engage concurrent neoantigen responses against tumors., Competing Interests: Declaration of interests T.J. is on the board of directors of Amgen and ThermoFisher Scientific, a co-founder of Dragonfly Therapeutics and T2 Biosystems, an SAB member of Dragonfly Therapeutics, SQZ Biotech, and Skyhawk Therapeutics and is president of Break Through Cancer. The T.J. lab receives funding from the Johnson & Johnson Lung Cancer Initiative and The Lustgarten Foundation for Pancreatic Cancer Research, but this funding did not support this research. A.R. is a founder and equity holder of Celsius Therapeutics, an equity holder in Immunitas Therapeutics and until August 31, 2020, was an SAB member of Syros Pharmaceuticals, Neogene Therapeutics, Asimov, and ThermoFisher Scientific. A.R. is an employee of Genentech (Roche Group) from August 1, 2020. S.S. is a consultant for RareCyte, Inc. A.M.C. interned at Skyhawk Therapeutics. None of these affiliations represent a conflict of interest with respect to the design or execution of this study or interpretation of data presented in this manuscript., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
15. Single-cell analyses identify circulating anti-tumor CD8 T cells and markers for their enrichment.
- Author
-
Pauken KE, Shahid O, Lagattuta KA, Mahuron KM, Luber JM, Lowe MM, Huang L, Delaney C, Long JM, Fung ME, Newcomer K, Tsai KK, Chow M, Guinn S, Kuchroo JR, Burke KP, Schenkel JM, Rosenblum MD, Daud AI, Sharpe AH, and Singer M
- Subjects
- Adenocarcinoma pathology, Animals, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Cell Line, Tumor, Colonic Neoplasms pathology, Female, Humans, Mice, Mice, Inbred C57BL, Programmed Cell Death 1 Receptor genetics, Programmed Cell Death 1 Receptor metabolism, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell metabolism, Transcriptome, Adenocarcinoma immunology, CD8-Positive T-Lymphocytes immunology, Colonic Neoplasms immunology, Melanoma blood, Melanoma immunology, Single-Cell Analysis methods, Skin Neoplasms blood, Skin Neoplasms immunology
- Abstract
The ability to monitor anti-tumor CD8+ T cell responses in the blood has tremendous therapeutic potential. Here, we used paired single-cell RNA and TCR sequencing to detect and characterize "tumor-matching" (TM) CD8+ T cells in the blood of mice with MC38 tumors or melanoma patients using the TCR as a molecular barcode. TM cells showed increased activation compared with nonmatching T cells in blood and were less exhausted than matching cells in tumors. Importantly, PD-1, which has been used to identify putative circulating anti-tumor CD8+ T cells, showed poor sensitivity for identifying TM cells. By leveraging the transcriptome, we identified candidate cell surface markers for TM cells in mice and patients and validated NKG2D, CD39, and CX3CR1 in mice. These data show that the TCR can be used to identify tumor-relevant cells for characterization, reveal unique transcriptional properties of TM cells, and develop marker panels for tracking and analysis of these cells., Competing Interests: Disclosures: K. Tsai reported personal fees from Regeneron, grants from Array/Pfizer, grants from Replimune, and grants from Oncosec outside the submitted work. M. Rosenblum reported other from TRex Bio and other from Sitryx Bio outside the submitted work. A. Daud reported grants from Merck, grants from BMS, other from Trex, grants from Pfizer, grants from Incyte, grants from Xencor, grants from Roche, and grants from Novartis during the conduct of the study as well as grants from Oncosec outside the submitted work. A. Sharpe reported grants from NIH R01 CA229851, grants from NIH U54 CA224088, grants from NIH P01 56299, and grants from NIH P01 39671 during the conduct of the study; personal fees from Surface Oncology, personal fees from Sqz Biotech, personal fees from Selecta, personal fees from Monopteros, personal fees from Elstar, personal fees from Elpiscience, grants from Novartis, grants from Roche, grants from Merck, grants from Ipsen, grants from UCB, and grants from Quark Ventures outside the submitted work. In addition, A. Sharpe had a patent number 7,432,059 with royalties paid (Roche, Merck, Bristol-Myers-Squibb, EMD-Serono, Boehringer-Ingelheim, AstraZeneca, Leica, Mayo Clinic, Dako and Novartis), a patent number 7,722,868 with royalties paid (Roche, Merck, Bristol-Myers-Squibb, EMD-Serono, Boehringer-Ingelheim, AstraZeneca, Leica, Mayo Clinic, Dako and Novartis), a patent number 8,652,465 licensed (Roche), a patent number 9,457,080 licensed (Roche), a patent number 9,683,048 licensed (Novartis), a patent number 9,815,898 licensed (Novartis), a patent number 9,845,356 licensed (Novartis), a patent number 10,202,454 licensed (Novartis), a patent number 10,457,733 licensed (Novartis), a patent number 9,580,684 issued (none), a patent number 9,988,452 issued (none), and a patent number 10,370,446 issued (none); A. Sharpe is on the scientific advisory boards for the Massachusetts General Cancer Center, Program in Cellular and Molecular Medicine at Boston Children's Hospital, and the Human Oncology and Pathogenesis Program at Memorial Sloan Kettering Cancer Center and is a scientific editor for the Journal of Experimental Medicine. M. Singer reported personal fees from Guardant Health outside the submitted work. No other disclosures were reported., (© 2021 Pauken et al.)
- Published
- 2021
- Full Text
- View/download PDF
16. Inhibitory signaling sustains a distinct early memory CD8 + T cell precursor that is resistant to DNA damage.
- Author
-
Johnnidis JB, Muroyama Y, Ngiow SF, Chen Z, Manne S, Cai Z, Song S, Platt JM, Schenkel JM, Abdel-Hakeem M, Beltra JC, Greenplate AR, Ali MA, Nzingha K, Giles JR, Harly C, Attanasio J, Pauken KE, Bengsch B, Paley MA, Tomov VT, Kurachi M, Vignali DAA, Sharpe AH, Reiner SL, Bhandoola A, Johnson FB, and Wherry EJ
- Subjects
- Animals, Antigens, CD genetics, CD8-Positive T-Lymphocytes metabolism, Cell Differentiation genetics, Cell Differentiation immunology, DNA Damage immunology, Disease Models, Animal, Female, Hepatocyte Nuclear Factor 1-alpha metabolism, Humans, Immunologic Memory genetics, Listeria monocytogenes immunology, Listeriosis microbiology, Lymphocyte Activation, Lymphocytic Choriomeningitis virology, Lymphocytic choriomeningitis virus immunology, Male, Memory T Cells metabolism, Mice, Mice, Knockout, Precursor Cells, T-Lymphoid metabolism, Programmed Cell Death 1 Receptor genetics, Lymphocyte Activation Gene 3 Protein, CD8-Positive T-Lymphocytes immunology, Listeriosis immunology, Lymphocytic Choriomeningitis immunology, Memory T Cells immunology, Precursor Cells, T-Lymphoid immunology
- Abstract
The developmental origins of memory T cells remain incompletely understood. During the expansion phase of acute viral infection, we identified a distinct subset of virus-specific CD8
+ T cells that possessed distinct characteristics including expression of CD62L, T cell factor 1 (TCF-1), and Eomesodermin; relative quiescence; expression of activation markers; and features of limited effector differentiation. These cells were a quantitatively minor subpopulation of the TCF-1+ pool and exhibited self-renewal, heightened DNA damage surveillance activity, and preferential long-term recall capacity. Despite features of memory and somewhat restrained proliferation during the expansion phase, this subset displayed evidence of stronger TCR signaling than other responding CD8+ T cells, coupled with elevated expression of multiple inhibitory receptors including programmed cell death 1 (PD-1), lymphocyte activating gene 3 (LAG-3), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), CD5, and CD160. Genetic ablation of PD-1 and LAG-3 compromised the formation of this CD62Lhi TCF-1+ subset and subsequent CD8+ T cell memory. Although central memory phenotype CD8+ T cells were formed in the absence of these cells, subsequent memory CD8+ T cell recall responses were compromised. Together, these results identify an important link between genome integrity maintenance and CD8+ T cell memory. Moreover, the data indicate a role for inhibitory receptors in preserving key memory CD8+ T cell precursors during initial activation and differentiation. Identification of this rare subpopulation within the memory CD8+ T cell precursor pool may help reconcile models of the developmental origin of long-term CD8+ T cell memory., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2021
- Full Text
- View/download PDF
17. Intratumoral injection of the seasonal flu shot converts immunologically cold tumors to hot and serves as an immunotherapy for cancer.
- Author
-
Newman JH, Chesson CB, Herzog NL, Bommareddy PK, Aspromonte SM, Pepe R, Estupinian R, Aboelatta MM, Buddhadev S, Tarabichi S, Lee M, Li S, Medina DJ, Giurini EF, Gupta KH, Guevara-Aleman G, Rossi M, Nowicki C, Abed A, Goldufsky JW, Broucek JR, Redondo RE, Rotter D, Jhawar SR, Wang SJ, Kohlhapp FJ, Kaufman HL, Thomas PG, Gupta V, Kuzel TM, Reiser J, Paras J, Kane MP, Singer EA, Malhotra J, Denzin LK, Sant'Angelo DB, Rabson AB, Lee LY, Lasfar A, Langenfeld J, Schenkel JM, Fidler MJ, Ruiz ES, Marzo AL, Rudra JS, Silk AW, and Zloza A
- Subjects
- Adjuvants, Immunologic administration & dosage, Animals, B-Lymphocytes, Basic-Leucine Zipper Transcription Factors genetics, CD8-Positive T-Lymphocytes immunology, Humans, Immunity, Cellular, Influenza A Virus, H1N1 Subtype immunology, Influenza, Human, Interleukin-10, Lung pathology, Lung Neoplasms drug therapy, Lung Neoplasms immunology, Lung Neoplasms pathology, Mice, Mice, Inbred C57BL, Repressor Proteins genetics, Seasons, Skin, Skin Neoplasms drug therapy, Skin Neoplasms pathology, Squalene administration & dosage, Tumor Microenvironment drug effects, Vaccination, Immunotherapy methods, Influenza Vaccines administration & dosage, Influenza Vaccines immunology, Influenza Vaccines therapeutic use, Injections, Intralesional, Neoplasms drug therapy, Neoplasms immunology
- Abstract
Reprogramming the tumor microenvironment to increase immune-mediated responses is currently of intense interest. Patients with immune-infiltrated "hot" tumors demonstrate higher treatment response rates and improved survival. However, only the minority of tumors are hot, and a limited proportion of patients benefit from immunotherapies. Innovative approaches that make tumors hot can have immediate impact particularly if they repurpose drugs with additional cancer-unrelated benefits. The seasonal influenza vaccine is recommended for all persons over 6 mo without prohibitive contraindications, including most cancer patients. Here, we report that unadjuvanted seasonal influenza vaccination via intratumoral, but not intramuscular, injection converts "cold" tumors to hot, generates systemic CD8
+ T cell-mediated antitumor immunity, and sensitizes resistant tumors to checkpoint blockade. Importantly, intratumoral vaccination also provides protection against subsequent active influenza virus lung infection. Surprisingly, a squalene-based adjuvanted vaccine maintains intratumoral regulatory B cells and fails to improve antitumor responses, even while protecting against active influenza virus lung infection. Adjuvant removal, B cell depletion, or IL-10 blockade recovers its antitumor effectiveness. Our findings propose that antipathogen vaccines may be utilized for both infection prevention and repurposing as a cancer immunotherapy., Competing Interests: Competing interest statement: P.K.B. and H.L.K. are employees of Replimune, Inc. E.A.S. receives research funding from Astellas/Medivation. M.J.F receives research funding from Pfizer and Biodesix, consulting funds from Astrazeneca and Pfizer, and honoraria from AstraZeneca, Merck, and Genentech. All other authors declare no conflicts of interest., (Copyright © 2020 the Author(s). Published by PNAS.)- Published
- 2020
- Full Text
- View/download PDF
18. CRISPR-mediated modeling and functional validation of candidate tumor suppressor genes in small cell lung cancer.
- Author
-
Ng SR, Rideout WM 3rd, Akama-Garren EH, Bhutkar A, Mercer KL, Schenkel JM, Bronson RT, and Jacks T
- Subjects
- Animals, Apoptosis genetics, CRISPR-Cas Systems genetics, Cell Line, Cell Proliferation genetics, Disease Models, Animal, Disease Progression, Feasibility Studies, Humans, Loss of Function Mutation, Lung pathology, Lung Neoplasms pathology, Mice, Mice, Transgenic, Neoplasm Staging, Retinoblastoma-Like Protein p107 genetics, Retinoblastoma-Like Protein p130 genetics, Small Cell Lung Carcinoma pathology, Tumor Burden genetics, Tumor Suppressor Protein p53 genetics, Gene Editing methods, Gene Expression Regulation, Neoplastic, Genes, Tumor Suppressor, Lung Neoplasms genetics, Small Cell Lung Carcinoma genetics
- Abstract
Small cell lung cancer (SCLC) is a highly aggressive subtype of lung cancer that remains among the most lethal of solid tumor malignancies. Recent genomic sequencing studies have identified many recurrently mutated genes in human SCLC tumors. However, the functional roles of most of these genes remain to be validated. Here, we have adapted the CRISPR-Cas9 system to a well-established murine model of SCLC to rapidly model loss-of-function mutations in candidate genes identified from SCLC sequencing studies. We show that loss of the gene p107 significantly accelerates tumor progression. Notably, compared with loss of the closely related gene p130 , loss of p107 results in fewer but larger tumors as well as earlier metastatic spread. In addition, we observe differences in proliferation and apoptosis as well as altered distribution of initiated tumors in the lung, resulting from loss of p107 or p130 Collectively, these data demonstrate the feasibility of using the CRISPR-Cas9 system to model loss of candidate tumor suppressor genes in SCLC, and we anticipate that this approach will facilitate efforts to investigate mechanisms driving tumor progression in this deadly disease., Competing Interests: Competing interest statement: T.J. is a member of the Board of Directors of Amgen and Thermo Fisher Scientific. He is also a co-founder of Dragonfly Therapeutics and T2 Biosystems. T.J. serves on the Scientific Advisory Board of Dragonfly Therapeutics, SQZ Biotech, and Skyhawk Therapeutics. None of these affiliations represent a conflict of interest with respect to the design or execution of this study or interpretation of data presented in this paper. Dr. Jacks’s laboratory currently also receives funding from the Johnson & Johnson Lung Cancer Initiative and Calico, but this funding did not support the research described in this paper., (Copyright © 2020 the Author(s). Published by PNAS.)
- Published
- 2020
- Full Text
- View/download PDF
19. IL-33 Signaling Alters Regulatory T Cell Diversity in Support of Tumor Development.
- Author
-
Li A, Herbst RH, Canner D, Schenkel JM, Smith OC, Kim JY, Hillman M, Bhutkar A, Cuoco MS, Rappazzo CG, Rogers P, Dang C, Jerby-Arnon L, Rozenblatt-Rosen O, Cong L, Birnbaum M, Regev A, and Jacks T
- Subjects
- Animals, CD8-Positive T-Lymphocytes immunology, Female, Immune Tolerance immunology, Immunosuppression Therapy methods, Male, Mice, Mice, Inbred C57BL, Neoplasms immunology, Tumor Microenvironment immunology, Interleukin-33 immunology, Signal Transduction immunology, T-Lymphocytes, Regulatory immunology
- Abstract
Regulatory T cells (T
regs ) can impair anti-tumor immune responses and are associated with poor prognosis in multiple cancer types. Tregs in human tumors span diverse transcriptional states distinct from those of peripheral Tregs , but their contribution to tumor development remains unknown. Here, we use single-cell RNA sequencing (RNA-seq) to longitudinally profile dynamic shifts in the distribution of Tregs in a genetically engineered mouse model of lung adenocarcinoma. In this model, interferon-responsive Tregs are more prevalent early in tumor development, whereas a specialized effector phenotype characterized by enhanced expression of the interleukin-33 receptor ST2 is predominant in advanced disease. Treg -specific deletion of ST2 alters the evolution of effector Treg diversity, increases infiltration of CD8+ T cells into tumors, and decreases tumor burden. Our study shows that ST2 plays a critical role in Treg -mediated immunosuppression in cancer, highlighting potential paths for therapeutic intervention., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
20. CD4 + resident memory T cells dominate immunosurveillance and orchestrate local recall responses.
- Author
-
Beura LK, Fares-Frederickson NJ, Steinert EM, Scott MC, Thompson EA, Fraser KA, Schenkel JM, Vezys V, and Masopust D
- Subjects
- Animals, Arenaviridae Infections virology, Cell Movement immunology, Chimera immunology, Female, Granzymes metabolism, Male, Mice, Mice, Inbred C57BL, Phenotype, RNA-Seq, Transcriptome, Arenaviridae Infections immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Immunologic Memory immunology, Immunologic Surveillance immunology, Lymphocytic choriomeningitis virus immunology
- Abstract
This study examines the extent to which memory CD4
+ T cells share immunosurveillance strategies with CD8+ resident memory T cells (TRM ). After acute viral infection, memory CD4+ T cells predominantly used residence to survey nonlymphoid tissues, albeit not as stringently as observed for CD8+ T cells. In contrast, memory CD4+ T cells were more likely to be resident within lymphoid organs than CD8+ T cells. Migration properties of memory-phenotype CD4+ T cells in non-SPF parabionts were similar, generalizing these results to diverse infections and conditions. CD4+ and CD8+ TRM shared overlapping transcriptional signatures and location-specific features, such as granzyme B expression in the small intestine, revealing tissue-specific and migration property-specific, in addition to lineage-specific, differentiation programs. Functionally, mucosal CD4+ TRM reactivation locally triggered both chemokine expression and broad immune cell activation. Thus, residence provides a dominant mechanism for regionalizing CD4+ T cell immunity, and location enforces shared transcriptional, phenotypic, and functional properties with CD8+ T cells., (© 2019 Beura et al.)- Published
- 2019
- Full Text
- View/download PDF
21. Use of a Blast Dominance-Hematogone Index for the Flow Cytometric Evaluation of Myelodysplastic Syndrome (MDS).
- Author
-
Schenkel JM, Hergott CB, Dudley G, Drew M, Charest K, and Dorfman DM
- Subjects
- Adult, Aged, Aged, 80 and over, CD13 Antigens analysis, Female, HLA-DR Antigens analysis, Humans, Male, Middle Aged, Myelodysplastic Syndromes pathology, Flow Cytometry methods, Myelodysplastic Syndromes immunology
- Abstract
Objectives: We tested whether combined flow cytometric assessment of loss of blast heterogeneity and decreased hematogones is a diagnostically useful approach for evaluation of myelodysplastic syndrome (MDS)., Methods: Bone marrow samples from patients with known MDS were analyzed by 10-color flow cytometric immunophenotyping and compared with normal bone marrow samples., Results: There was loss of blast heterogeneity in patients with MDS compared with normal bone marrow samples, based on the relative size of the dominant blast population (83.0% vs 64.8%) and fewer hematogones (0.08% vs 1.39%). The size of the largest blast population divided by the fraction of hematogones (blast dominance-hematogone [BDH] index) was significantly larger in MDS compared with normal cases (27,084 vs 190, P < .0001; receiver operating characteristic area under the curve = 0.96)., Conclusions: The BDH index is more sensitive and specific than loss of blast heterogeneity or decrease in hematogones for detecting MDS in bone marrow samples and may be useful in clinical practice., (© American Society for Clinical Pathology, 2019. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
22. Plateletpheresis-associated lymphopenia in frequent platelet donors.
- Author
-
Gansner JM, Rahmani M, Jonsson AH, Fortin BM, Brimah I, Ellis M, Smeland-Wagman R, Li ZJ, Schenkel JM, Brenner MB, Yefidoff-Freedman R, Sloan SR, Berliner N, Issa NC, Baden LR, Longo DL, Wesemann DR, Neuberg D, Rao DA, and Kaufman RM
- Subjects
- Adult, Aged, Cross-Sectional Studies, Female, Humans, Male, Middle Aged, Platelet Count, Prognosis, Young Adult, Blood Donors statistics & numerical data, Blood Platelets cytology, Lymphopenia etiology, Plateletpheresis adverse effects
- Abstract
More than 1 million apheresis platelet collections are performed annually in the United States. After 2 healthy plateletpheresis donors were incidentally found to have low CD4
+ T-lymphocyte counts, we investigated whether plateletpheresis causes lymphopenia. We conducted a cross-sectional single-center study of platelet donors undergoing plateletpheresis with the Trima Accel, which removes leukocytes continuously with its leukoreduction system chamber. We recruited 3 groups of platelet donors based on the total number of plateletpheresis sessions in the prior 365 days: 1 or 2, 3 to 19, or 20 to 24. CD4+ T-lymphocyte counts were <200 cells per microliter in 0/20, 2/20, and 6/20 donors, respectively ( P = .019), and CD8+ T-lymphocyte counts were low in 0/20, 4/20, and 11/20 donors, respectively ( P < .001). The leukoreduction system chamber's lymphocyte-extraction efficiency was ∼15% to 20% for all groups. Immunophenotyping showed decreases in naive CD4+ T-lymphocyte and T helper 17 (Th17) cell percentages, increases in CD4+ and CD8+ effector memory, Th1, and regulatory T cell percentages, and stable naive CD8+ and Th2 percentages across groups. T-cell receptor repertoire analyses showed similar clonal diversity in all groups. Donor screening questionnaires supported the good health of the donors, who tested negative at each donation for multiple pathogens, including HIV. Frequent plateletpheresis utilizing a leukoreduction system chamber is associated with CD4+ and CD8+ T-cell lymphopenia in healthy platelet donors. The mechanism may be repeated extraction of these cells during plateletpheresis. The cytopenias do not appear to be harmful., (© 2019 by The American Society of Hematology.)- Published
- 2019
- Full Text
- View/download PDF
23. A phase II trial of riluzole, an antagonist of metabotropic glutamate receptor 1 (GRM1) signaling, in patients with advanced melanoma.
- Author
-
Mehnert JM, Silk AW, Lee JH, Dudek L, Jeong BS, Li J, Schenkel JM, Sadimin E, Kane M, Lin H, Shih WJ, Zloza A, Chen S, and Goydos JS
- Subjects
- Adult, Female, Humans, Male, Middle Aged, Receptors, Metabotropic Glutamate biosynthesis, Riluzole adverse effects, Down-Regulation drug effects, MAP Kinase Signaling System drug effects, Melanoma drug therapy, Melanoma metabolism, Melanoma pathology, Receptors, Metabotropic Glutamate antagonists & inhibitors, Riluzole administration & dosage
- Abstract
Studies demonstrate that GRM, expressed by >60% of human melanomas, may be a therapeutic target. We performed a phase II trial of 100 mg PO bid of riluzole, an inhibitor of GRM1 signaling, in patients with advanced melanoma with the primary endpoint of response rate. Thirteen patients with GRM1-positive tumors were enrolled. No objective responses were observed, and accrual was stopped. Stable disease was noted in six (46%) patients, with one patient on study for 42 weeks. Riluzole was well tolerated, with fatigue (62%) as the most common adverse event. Downregulation of MAPK and PI3K/AKT was noted in 33% of paired tumor biopsies. Hypothesis-generating correlative studies suggested that downregulation of angiogenic markers and increased leukocytes at the active edge of tumor correlate with clinical benefit. Pharmacokinetic analysis showed interpatient variability consistent with prior riluzole studies. Future investigations should interrogate mechanisms of biologic activity and advance the development of agents with improved bioavailability., (© 2018 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
24. T Cells in Nonlymphoid Tissues Give Rise to Lymph-Node-Resident Memory T Cells.
- Author
-
Beura LK, Wijeyesinghe S, Thompson EA, Macchietto MG, Rosato PC, Pierson MJ, Schenkel JM, Mitchell JS, Vezys V, Fife BT, Shen S, and Masopust D
- Subjects
- Animals, Antigens, CD analysis, Antigens, Differentiation, T-Lymphocyte analysis, Female, Lectins, C-Type analysis, Lymphocytic Choriomeningitis immunology, Mice, Mice, Inbred C57BL, CD8-Positive T-Lymphocytes immunology, Immunologic Memory immunology, Lymph Nodes immunology
- Abstract
Immunosurveillance of secondary lymphoid organs (SLO) is performed by central memory T cells that recirculate through blood. Resident memory T (Trm) cells remain parked in nonlymphoid tissues and often stably express CD69. We recently identified Trm cells within SLO, but the origin and phenotype of these cells remains unclear. Using parabiosis of "dirty" mice, we found that CD69 expression is insufficient to infer stable residence of SLO Trm cells. Restimulation of nonlymphoid memory CD8
+ T cells within the skin or mucosa resulted in a substantial increase in bona fide Trm cells specifically within draining lymph nodes. SLO Trm cells derived from emigrants from nonlymphoid tissues and shared some transcriptional and phenotypic signatures associated with nonlymphoid Trm cells. These data indicate that nonlymphoid cells can give rise to SLO Trm cells and suggest vaccination strategies by which memory CD8+ T cell immunosurveillance can be regionalized to specific lymph nodes., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
25. Intravital mucosal imaging of CD8 + resident memory T cells shows tissue-autonomous recall responses that amplify secondary memory.
- Author
-
Beura LK, Mitchell JS, Thompson EA, Schenkel JM, Mohammed J, Wijeyesinghe S, Fonseca R, Burbach BJ, Hickman HD, Vezys V, Fife BT, and Masopust D
- Subjects
- Animals, Female, Intravital Microscopy, Mice, Mucous Membrane cytology, Skin immunology, CD8-Positive T-Lymphocytes immunology, Immunity, Mucosal immunology, Immunologic Memory immunology, Immunologic Surveillance immunology, Mucous Membrane immunology
- Abstract
CD8
+ T cell immunosurveillance dynamics influence the outcome of intracellular infections and cancer. Here we used two-photon intravital microscopy to visualize the responses of CD8+ resident memory T cells (TRM cells) within the reproductive tracts of live female mice. We found that mucosal TRM cells were highly motile, but paused and underwent in situ division after local antigen challenge. TRM cell reactivation triggered the recruitment of recirculating memory T cells that underwent antigen-independent TRM cell differentiation in situ. However, the proliferation of pre-existing TRM cells dominated the local mucosal recall response and contributed most substantially to the boosted secondary TRM cell population. We observed similar results in skin. Thus, TRM cells can autonomously regulate the expansion of local immunosurveillance independently of central memory or proliferation in lymphoid tissue.- Published
- 2018
- Full Text
- View/download PDF
26. Oncolytic Viruses-Natural and Genetically Engineered Cancer Immunotherapies.
- Author
-
Jhawar SR, Thandoni A, Bommareddy PK, Hassan S, Kohlhapp FJ, Goyal S, Schenkel JM, Silk AW, and Zloza A
- Abstract
There has long been interest in innovating an approach by which tumor cells can be selectively and specifically targeted and destroyed. The discovery of viruses that lyse tumor cells, termed oncolytic viruses (OVs), has led to a revolution in the treatment of cancer. The potential of OVs to improve the therapeutic ratio is derived from their ability to preferentially infect and replicate in cancer cells while avoiding destruction of normal cells surrounding the tumor. Two main mechanisms exist through which these viruses are reported to improve outcomes: direct lysis of tumor cells and indirect augmentation of host anti-tumor immunity. With these factors in mind, viruses are chosen or modified to selectively target tumor cells, decrease pathogenicity to normal cells, decrease the antiviral immune response (to prevent viral clearance), and increase the antitumor immune response. While only one OV has been approved for the treatment of cancer in the United States, and only two other OVs have been approved worldwide, a wide spectrum of OVs are in various stages of preclinical development and in clinical trials. These viruses are being studied as alternatives and adjuncts to more traditional cancer therapies including surgical resection, chemotherapy, radiation, hormonal therapies, targeted therapies, and other immunotherapies. Here, we review the natural characteristics and genetically engineered modifications that enhance the effectiveness of OVs for the treatment of cancer.
- Published
- 2017
- Full Text
- View/download PDF
27. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response.
- Author
-
Huang AC, Postow MA, Orlowski RJ, Mick R, Bengsch B, Manne S, Xu W, Harmon S, Giles JR, Wenz B, Adamow M, Kuk D, Panageas KS, Carrera C, Wong P, Quagliarello F, Wubbenhorst B, D'Andrea K, Pauken KE, Herati RS, Staupe RP, Schenkel JM, McGettigan S, Kothari S, George SM, Vonderheide RH, Amaravadi RK, Karakousis GC, Schuchter LM, Xu X, Nathanson KL, Wolchok JD, Gangadhar TC, and Wherry EJ
- Subjects
- Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized immunology, Antibodies, Monoclonal, Humanized pharmacokinetics, Antibodies, Monoclonal, Humanized therapeutic use, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes metabolism, Female, Humans, Ki-67 Antigen immunology, Ki-67 Antigen metabolism, Male, Melanoma blood supply, Melanoma pathology, Neoplasm Staging, Phenotype, Treatment Outcome, CD8-Positive T-Lymphocytes immunology, Melanoma drug therapy, Melanoma immunology, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor immunology, Tumor Burden immunology
- Abstract
Despite the success of monotherapies based on blockade of programmed cell death 1 (PD-1) in human melanoma, most patients do not experience durable clinical benefit. Pre-existing T-cell infiltration and/or the presence of PD-L1 in tumours may be used as indicators of clinical response; however, blood-based profiling to understand the mechanisms of PD-1 blockade has not been widely explored. Here we use immune profiling of peripheral blood from patients with stage IV melanoma before and after treatment with the PD-1-targeting antibody pembrolizumab and identify pharmacodynamic changes in circulating exhausted-phenotype CD8 T cells (T
ex cells). Most of the patients demonstrated an immunological response to pembrolizumab. Clinical failure in many patients was not solely due to an inability to induce immune reinvigoration, but rather resulted from an imbalance between T-cell reinvigoration and tumour burden. The magnitude of reinvigoration of circulating Tex cells determined in relation to pretreatment tumour burden correlated with clinical response. By focused profiling of a mechanistically relevant circulating T-cell subpopulation calibrated to pretreatment disease burden, we identify a clinically accessible potential on-treatment predictor of response to PD-1 blockade.- Published
- 2017
- Full Text
- View/download PDF
28. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade.
- Author
-
Pauken KE, Sammons MA, Odorizzi PM, Manne S, Godec J, Khan O, Drake AM, Chen Z, Sen DR, Kurachi M, Barnitz RA, Bartman C, Bengsch B, Huang AC, Schenkel JM, Vahedi G, Haining WN, Berger SL, and Wherry EJ
- Subjects
- Animals, B7-H1 Antigen antagonists & inhibitors, CD8-Positive T-Lymphocytes transplantation, Cell Lineage genetics, Cellular Reprogramming immunology, Female, Gene Regulatory Networks, Immunotherapy, Interleukin-7 metabolism, Mice, Mice, Inbred C57BL, Transcription, Genetic, B7-H1 Antigen genetics, CD8-Positive T-Lymphocytes immunology, Cellular Reprogramming genetics, Epigenesis, Genetic, Immunologic Memory genetics
- Abstract
Blocking Programmed Death-1 (PD-1) can reinvigorate exhausted CD8 T cells (T
EX ) and improve control of chronic infections and cancer. However, whether blocking PD-1 can reprogram TEX into durable memory T cells (TMEM ) is unclear. We found that reinvigoration of TEX in mice by PD-L1 blockade caused minimal memory development. After blockade, reinvigorated TEX became reexhausted if antigen concentration remained high and failed to become TMEM upon antigen clearance. TEX acquired an epigenetic profile distinct from that of effector T cells (TEFF ) and TMEM cells that was minimally remodeled after PD-L1 blockade. This finding suggests that TEX are a distinct lineage of CD8 T cells. Nevertheless, PD-1 pathway blockade resulted in transcriptional rewiring and reengagement of effector circuitry in the TEX epigenetic landscape. These data indicate that epigenetic fate inflexibility may limit current immunotherapies., (Copyright © 2016, American Association for the Advancement of Science.)- Published
- 2016
- Full Text
- View/download PDF
29. Non-oncogenic Acute Viral Infections Disrupt Anti-cancer Responses and Lead to Accelerated Cancer-Specific Host Death.
- Author
-
Kohlhapp FJ, Huelsmann EJ, Lacek AT, Schenkel JM, Lusciks J, Broucek JR, Goldufsky JW, Hughes T, Zayas JP, Dolubizno H, Sowell RT, Kühner R, Burd S, Kubasiak JC, Nabatiyan A, Marshall S, Bommareddy PK, Li S, Newman JH, Monken CE, Shafikhani SH, Marzo AL, Guevara-Patino JA, Lasfar A, Thomas PG, Lattime EC, Kaufman HL, and Zloza A
- Subjects
- Acute Disease, Animals, CD8-Positive T-Lymphocytes immunology, Cell Proliferation, Lung pathology, Mice, Inbred BALB C, Mice, Inbred C57BL, Programmed Cell Death 1 Receptor metabolism, Melanoma immunology, Melanoma pathology, Oncogenes, Orthomyxoviridae Infections pathology
- Abstract
In light of increased cancer prevalence and cancer-specific deaths in patients with infections, we investigated whether infections alter anti-tumor immune responses. We report that acute influenza infection of the lung promotes distal melanoma growth in the dermis and leads to accelerated cancer-specific host death. Furthermore, we show that during influenza infection, anti-melanoma CD8
+ T cells are shunted from the tumor to the infection site, where they express high levels of the inhibitory receptor programmed cell death protein 1 (PD-1). Immunotherapy to block PD-1 reverses this loss of anti-tumor CD8+ T cells from the tumor and decreases infection-induced tumor growth. Our findings show that acute non-oncogenic infection can promote cancer growth, raising concerns regarding acute viral illness sequelae. They also suggest an unexpected role for PD-1 blockade in cancer immunotherapy and provide insight into the immune response when faced with concomitant challenges., (Copyright © 2016 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2016
- Full Text
- View/download PDF
30. Heterologous Vaccination and Checkpoint Blockade Synergize To Induce Antileukemia Immunity.
- Author
-
Manlove LS, Schenkel JM, Manlove KR, Pauken KE, Williams RT, Vezys V, and Farrar MA
- Subjects
- Animals, Disease Models, Animal, Mice, Mice, Inbred C57BL, Mice, Knockout, Cell Cycle Checkpoints physiology, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma immunology, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Vaccination
- Abstract
Checkpoint blockade-based immunotherapies are effective in cancers with high numbers of nonsynonymous mutations. In contrast, current paradigms suggest that such approaches will be ineffective in cancers with few nonsynonymous mutations. To examine this issue, we made use of a murine model of BCR-ABL(+) B-lineage acute lymphoblastic leukemia. Using a principal component analysis, we found that robust MHC class II expression, coupled with appropriate costimulation, correlated with lower leukemic burden. We next assessed whether checkpoint blockade or therapeutic vaccination could improve survival in mice with pre-established leukemia. Consistent with the low mutation load in our leukemia model, we found that checkpoint blockade alone had only modest effects on survival. In contrast, robust heterologous vaccination with a peptide derived from the BCR-ABL fusion (BAp), a key driver mutation, generated a small population of mice that survived long-term. Checkpoint blockade strongly synergized with heterologous vaccination to enhance overall survival in mice with leukemia. Enhanced survival did not correlate with numbers of BAp:I-A(b)-specific T cells, but rather with increased expression of IL-10, IL-17, and granzyme B and decreased expression of programmed death 1 on these cells. Our findings demonstrate that vaccination to key driver mutations cooperates with checkpoint blockade and allows for immune control of cancers with low nonsynonymous mutation loads., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
31. IL-15-Independent Maintenance of Tissue-Resident and Boosted Effector Memory CD8 T Cells.
- Author
-
Schenkel JM, Fraser KA, Casey KA, Beura LK, Pauken KE, Vezys V, and Masopust D
- Subjects
- Animals, Cell Proliferation, Cells, Cultured, Cytotoxicity, Immunologic, Homeostasis, Immunization, Secondary, Mice, Mice, Inbred C57BL, Viral Vaccines immunology, CD8-Positive T-Lymphocytes immunology, Immunologic Memory, Interleukin-15 metabolism, Mucous Membrane immunology, T-Lymphocyte Subsets immunology
- Abstract
IL-15 regulates central and effector memory CD8 T cell (TCM and TEM, respectively) homeostatic proliferation, maintenance, and longevity. Consequently, IL-15 availability hypothetically defines the carrying capacity for total memory CD8 T cells within the host. In conflict with this hypothesis, previous observations demonstrated that boosting generates preternaturally abundant TEM that increases the total quantity of memory CD8 T cells in mice. In this article, we provide a potential mechanistic explanation by reporting that boosted circulating TEM do not require IL-15 for maintenance. We also investigated tissue-resident memory CD8 T cells (TRM), which protect nonlymphoid tissues from reinfection. We observed up to a 50-fold increase in the total magnitude of TRM in mouse mucosal tissues after boosting, suggesting that the memory T cell capacity in tissues is flexible and that TRM may not be under the same homeostatic regulation as primary central memory CD8 T cells and TEM Further analysis identified distinct TRM populations that depended on IL-15 for homeostatic proliferation and survival, depended on IL-15 for homeostatic proliferation but not for survival, or did not depend on IL-15 for either process. These observations on the numerical regulation of T cell memory indicate that there may be significant heterogeneity among distinct TRM populations and also argue against the common perception that developing vaccines that confer protection by establishing abundant TEM and TRM will necessarily erode immunity to previously encountered pathogens as the result of competition for IL-15., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
32. Normalizing the environment recapitulates adult human immune traits in laboratory mice.
- Author
-
Beura LK, Hamilton SE, Bi K, Schenkel JM, Odumade OA, Casey KA, Thompson EA, Fraser KA, Rosato PC, Filali-Mouhim A, Sekaly RP, Jenkins MK, Vezys V, Haining WN, Jameson SC, and Masopust D
- Subjects
- Adult, Animals, Cell Differentiation, Environmental Exposure, Female, Humans, Immunity, Innate immunology, Immunologic Memory, Infant, Newborn, Male, Mice, Phenotype, Specific Pathogen-Free Organisms, T-Lymphocytes cytology, T-Lymphocytes immunology, Virus Diseases immunology, Virus Diseases virology, Animal Husbandry methods, Animals, Laboratory immunology, Animals, Wild immunology, Environment, Immune System immunology, Immunity immunology, Models, Animal
- Abstract
Our current understanding of immunology was largely defined in laboratory mice, partly because they are inbred and genetically homogeneous, can be genetically manipulated, allow kinetic tissue analyses to be carried out from the onset of disease, and permit the use of tractable disease models. Comparably reductionist experiments are neither technically nor ethically possible in humans. However, there is growing concern that laboratory mice do not reflect relevant aspects of the human immune system, which may account for failures to translate disease treatments from bench to bedside. Laboratory mice live in abnormally hygienic specific pathogen free (SPF) barrier facilities. Here we show that standard laboratory mouse husbandry has profound effects on the immune system and that environmental changes produce mice with immune systems closer to those of adult humans. Laboratory mice--like newborn, but not adult, humans--lack effector-differentiated and mucosally distributed memory T cells. These cell populations were present in free-living barn populations of feral mice and pet store mice with diverse microbial experience, and were induced in laboratory mice after co-housing with pet store mice, suggesting that the environment is involved in the induction of these cells. Altering the living conditions of mice profoundly affected the cellular composition of the innate and adaptive immune systems, resulted in global changes in blood cell gene expression to patterns that more closely reflected the immune signatures of adult humans rather than neonates, altered resistance to infection, and influenced T-cell differentiation in response to a de novo viral infection. These data highlight the effects of environment on the basal immune state and response to infection and suggest that restoring physiological microbial exposure in laboratory mice could provide a relevant tool for modelling immunological events in free-living organisms, including humans.
- Published
- 2016
- Full Text
- View/download PDF
33. Interleukin-2-Dependent Allergen-Specific Tissue-Resident Memory Cells Drive Asthma.
- Author
-
Hondowicz BD, An D, Schenkel JM, Kim KS, Steach HR, Krishnamurty AT, Keitany GJ, Garza EN, Fraser KA, Moon JJ, Altemeier WA, Masopust D, and Pepper M
- Subjects
- Allergens immunology, Animals, Antigens, Dermatophagoides immunology, Cell Differentiation immunology, Cell Separation, Disease Models, Animal, Female, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Pyroglyphidae immunology, Asthma immunology, Immunologic Memory immunology, Interleukin-2 immunology, Lung immunology, Th2 Cells immunology
- Abstract
Exposure to inhaled allergens generates T helper 2 (Th2) CD4(+) T cells that contribute to episodes of inflammation associated with asthma. Little is known about allergen-specific Th2 memory cells and their contribution to airway inflammation. We generated reagents to understand how endogenous CD4(+) T cells specific for a house dust mite (HDM) allergen form and function. After allergen exposure, HDM-specific memory cells persisted as central memory cells in the lymphoid organs and tissue-resident memory cells in the lung. Experimental blockade of lymphocyte migration demonstrated that lung-resident cells were sufficient to induce airway hyper-responsiveness, which depended upon CD4(+) T cells. Investigation into the differentiation of pathogenic Trm cells revealed that interleukin-2 (IL-2) signaling was required for residency and directed a program of tissue homing migrational cues. These studies thus identify IL-2-dependent resident Th2 memory cells as drivers of lung allergic responses., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
34. NK cells and CD8+ T cells cooperate to improve therapeutic responses in melanoma treated with interleukin-2 (IL-2) and CTLA-4 blockade.
- Author
-
Kohlhapp FJ, Broucek JR, Hughes T, Huelsmann EJ, Lusciks J, Zayas JP, Dolubizno H, Fleetwood VA, Grin A, Hill GE, Poshepny JL, Nabatiyan A, Ruby CE, Snook JD, Rudra JS, Schenkel JM, Masopust D, Zloza A, and Kaufman HL
- Abstract
Background: Melanoma is one of the few types of cancer with an increasing annual incidence. While a number of immunotherapies for melanoma have been associated with significant clinical benefit, including high-dose IL-2 and cytotoxic T lymphocyte antigen 4 (CTLA-4) blockade, clinical response to either of these single agents has been limited to 11-20% of treated patients. Therefore, in this study, we sought to test the hypothesis that the combination of IL-2 and CTLA-4 blockade could mediate a more profound therapeutic response., Methods: Here, B6 mice were challenged with poorly immunogenic B16 melanoma on day 0, and treated with CTLA-4 blocking antibody (100 μg/mouse) on days 3, 6, and 9, and IL-2 (100,000 units) twice daily on days 4-8, or both., Results: A highly significant synergistic effect that delayed tumor growth and prolonged survival was demonstrated with the combination immunotherapy compared to either monotherapy alone. The therapeutic effect of combination immunotherapy was dependent on both CD8+ T and NK cells and co-depletion of these subsets (but not either one alone) abrogated the therapeutic effect. CTLA-4 blockade increased immune cell infiltration (including CD8+ T cells and NK cells) in the tumor and IL-2 reduced the proportion of highly differentiated/exhausted tumor-infiltrating NK cells., Conclusions: These results have implications for the design of clinical trials in patients with metastatic melanoma and provide new insights into how the immune system may be mediating anti-tumor activity with combination IL-2 and CTLA-4 blockade in melanoma.
- Published
- 2015
- Full Text
- View/download PDF
35. Quantifying Memory CD8 T Cells Reveals Regionalization of Immunosurveillance.
- Author
-
Steinert EM, Schenkel JM, Fraser KA, Beura LK, Manlove LS, Igyártó BZ, Southern PJ, and Masopust D
- Subjects
- Animals, Antigens, CD metabolism, Antigens, Differentiation, T-Lymphocyte metabolism, CD8-Positive T-Lymphocytes immunology, Cell Movement, Inflammation immunology, Lectins, C-Type metabolism, Mice, Mice, Inbred C57BL, Arenaviridae Infections immunology, Immunologic Memory, Lymphocytic choriomeningitis virus physiology, Monitoring, Immunologic, T-Lymphocyte Subsets immunology
- Abstract
Memory CD8 T cells protect against intracellular pathogens by scanning host cell surfaces; thus, infection detection rates depend on memory cell number and distribution. Population analyses rely on cell isolation from whole organs, and interpretation is predicated on presumptions of near complete cell recovery. Paradigmatically, memory is parsed into central, effector, and resident subsets, ostensibly defined by immunosurveillance patterns but in practice identified by phenotypic markers. Because isolation methods ultimately inform models of memory T cell differentiation, protection, and vaccine translation, we tested their validity via parabiosis and quantitative immunofluorescence microscopy of a mouse memory CD8 T cell population. We report three major findings: lymphocyte isolation fails to recover most cells and biases against certain subsets, residents greatly outnumber recirculating cells within non-lymphoid tissues, and memory subset homing to inflammation does not conform to previously hypothesized migration patterns. These results indicate that most host cells are surveyed for reinfection by segregated residents rather than by recirculating cells that migrate throughout the blood and body., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
36. Cutting edge: identification of autoreactive CD4+ and CD8+ T cell subsets resistant to PD-1 pathway blockade.
- Author
-
Pauken KE, Nelson CE, Martinov T, Spanier JA, Heffernan JR, Sahli NL, Quarnstrom CF, Osum KC, Schenkel JM, Jenkins MK, Blazar BR, Vezys V, and Fife BT
- Subjects
- Animals, Autoimmune Diseases genetics, Autoimmune Diseases pathology, B7-H1 Antigen genetics, B7-H1 Antigen immunology, CD4-Positive T-Lymphocytes pathology, CD8-Positive T-Lymphocytes pathology, Cell Differentiation genetics, Cell Differentiation immunology, Disease Models, Animal, Disease Susceptibility immunology, Disease Susceptibility pathology, Female, Immune Tolerance genetics, Mice, Mice, Inbred NOD, Mice, Knockout, Programmed Cell Death 1 Receptor genetics, Signal Transduction genetics, Autoimmune Diseases immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Clonal Anergy, Programmed Cell Death 1 Receptor immunology, Signal Transduction immunology
- Abstract
Programmed death-1 (PD-1) promotes T cell tolerance. Despite therapeutically targeting this pathway for chronic infections and tumors, little is known about how different T cell subsets are affected during blockade. We examined PD-1/PD ligand 1 (PD-L1) regulation of self-antigen-specific CD4 and CD8 T cells in autoimmune-susceptible models. PD-L1 blockade increased insulin-specific effector CD4 T cells in type 1 diabetes. However, anergic islet-specific CD4 T cells were resistant to PD-L1 blockade. Additionally, PD-L1 was critical for induction, but not maintenance, of CD8 T cell intestinal tolerance. PD-L1 blockade enhanced functionality of effector T cells, whereas established tolerant or anergic T cells were not dependent on PD-1/PD-L1 signaling to remain unresponsive. This highlights the existence of Ag-experienced T cell subsets that do not rely on PD-1/PD-L1 regulation. These findings illustrate how positive treatment outcomes and autoimmunity development during PD-1/PD-L1 inhibition are linked to the differentiation state of a T cell., (Copyright © 2015 by The American Association of Immunologists, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
37. Lymphocytic choriomeningitis virus persistence promotes effector-like memory differentiation and enhances mucosal T cell distribution.
- Author
-
Beura LK, Anderson KG, Schenkel JM, Locquiao JJ, Fraser KA, Vezys V, Pepper M, and Masopust D
- Subjects
- Animals, Antigens, CD immunology, Antigens, Differentiation, T-Lymphocyte immunology, CD4-Positive T-Lymphocytes pathology, CD8-Positive T-Lymphocytes pathology, Cell Movement immunology, Female, Gene Expression Regulation immunology, Granzymes immunology, Integrin alpha Chains immunology, Integrin alpha4 immunology, Integrin beta Chains immunology, Intestinal Mucosa pathology, Lectins, C-Type immunology, Lymphocytic Choriomeningitis pathology, Mice, Organ Specificity immunology, Receptors, CXCR4 immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Immunity, Mucosal, Immunologic Memory, Intestinal Mucosa immunology, Lymphocytic Choriomeningitis immunology, Lymphocytic choriomeningitis virus immunology
- Abstract
Vaccines are desired that maintain abundant memory T cells at nonlymphoid sites of microbial exposure, where they may be anatomically positioned for immediate pathogen interception. Here, we test the impact of antigen persistence on mouse CD8 and CD4 T cell distribution and differentiation by comparing responses to infections with different strains of LCMV that cause either acute or chronic infections. We used in vivo labeling techniques that discriminate between T cells present within tissues and abundant populations that fail to be removed from vascular compartments, despite perfusion. LCMV persistence caused up to ∼30-fold more virus-specific CD8 T cells to distribute to the lung compared with acute infection. Persistent infection also maintained mucosal-homing α4β7 integrin expression, higher granzyme B expression, alterations in the expression of the TRM markers CD69 and CD103, and greater accumulation of virus-specific CD8 T cells in the large intestine, liver, kidney, and female reproductive tract. Persistent infection also increased LCMV-specific CD4 T cell quantity in mucosal tissues and induced maintenance of CXCR4, an HIV coreceptor. This study clarifies the relationship between viral persistence and CD4 and CD8 T cell distribution and mucosal phenotype, indicating that chronic LCMV infection magnifies T cell migration to nonlymphoid tissues., (© Society for Leukocyte Biology.)
- Published
- 2015
- Full Text
- View/download PDF
38. Tissue-resident memory T cells.
- Author
-
Schenkel JM and Masopust D
- Subjects
- Animals, Cell Differentiation, Cell Movement, Humans, Immunity, Immunologic Memory, T-Lymphocyte Subsets immunology, T-Lymphocytes immunology
- Abstract
Tissue-resident memory T (Trm) cells constitute a recently identified lymphocyte lineage that occupies tissues without recirculating. They provide a first response against infections reencountered at body surfaces, where they accelerate pathogen clearance. Because Trm cells are not present within peripheral blood, they have not yet been well characterized, but are transcriptionally, phenotypically, and functionally distinct from recirculating central and effector memory T cells. In this review, we will summarize current knowledge of Trm cell ontogeny, regulation, maintenance, and function and will highlight technical considerations for studying this population., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
39. High-dose IL-2 induces rapid albumin uptake by endothelial cells through Src-dependent caveolae-mediated endocytosis.
- Author
-
Zloza A, Kim DW, Broucek J, Schenkel JM, and Kaufman HL
- Subjects
- Capillary Permeability drug effects, Caveolae metabolism, Cells, Cultured, Endocytosis drug effects, Endothelial Cells metabolism, Humans, Interleukin-2 adverse effects, Signal Transduction, src-Family Kinases metabolism, Albumins metabolism, Caveolin 1 metabolism, Endothelial Cells drug effects, Interleukin-2 pharmacology
- Abstract
High-dose interleukin-2 (HDIL2) treatment of patients with metastatic melanoma and renal cell carcinoma is associated with durable responses, but therapy is accompanied by significant toxicity related to vascular leak syndrome (VLS). Currently, the cause of VLS is not well defined; however, based on the role of endothelial cell (EC) permeability in VLS and the commonly observed hypoalbuminemia in patients receiving HDIL2 therapy, we established an in vitro approach utilizing primary human pulmonary microvascular ECs to monitor the effect of HDIL2 therapy on albumin uptake. We found that HDIL2 treatment of ECs results in albumin colocalization with caveolin-1 leading to albumin uptake by ECs. This albumin uptake occurs through caveolae-mediated but not clathrin-mediated endocytosis and is abrogated with inhibition of the Src tyrosine kinase pathway. These findings provide insight into how IL-2 induces VLS and may help identify potential targets for prevention of toxicity without affecting the therapeutic activity of HDIL2.
- Published
- 2014
- Full Text
- View/download PDF
40. T cell memory. Resident memory CD8 T cells trigger protective innate and adaptive immune responses.
- Author
-
Schenkel JM, Fraser KA, Beura LK, Pauken KE, Vezys V, and Masopust D
- Subjects
- Animals, Antigens, Viral immunology, Female, Immunity, Humoral immunology, Interferon-gamma immunology, Mice, Mice, Inbred C57BL, Mucous Membrane immunology, Mucous Membrane virology, Vascular Cell Adhesion Molecule-1 immunology, Adaptive Immunity immunology, CD8-Positive T-Lymphocytes immunology, Immunity, Innate immunology, Immunologic Memory, Virus Diseases immunology
- Abstract
The pathogen recognition theory dictates that, upon viral infection, the innate immune system first detects microbial products and then responds by providing instructions to adaptive CD8 T cells. Here, we show in mice that tissue resident memory CD8 T cells (T(RM) cells), non-recirculating cells located at common sites of infection, can achieve near-sterilizing immunity against viral infections by reversing this flow of information. Upon antigen resensitization within the mouse female reproductive mucosae, CD8(+) T(RM) cells secrete cytokines that trigger rapid adaptive and innate immune responses, including local humoral responses, maturation of local dendritic cells, and activation of natural killer cells. This provided near-sterilizing immunity against an antigenically unrelated viral infection. Thus, CD8(+) T(RM) cells rapidly trigger an antiviral state by amplifying receptor-derived signals from previously encountered pathogens., (Copyright © 2014, American Association for the Advancement of Science.)
- Published
- 2014
- Full Text
- View/download PDF
41. Interleukin-2 alters distribution of CD144 (VE-cadherin) in endothelial cells.
- Author
-
Kim DW, Zloza A, Broucek J, Schenkel JM, Ruby C, Samaha G, and Kaufman HL
- Subjects
- Adult, Aged, Base Sequence, CD146 Antigen metabolism, Cells, Cultured, DNA Primers, Endothelial Cells cytology, Female, Humans, Male, Middle Aged, Polymerase Chain Reaction, Endothelial Cells immunology, Interleukin-2 physiology
- Abstract
Background: High-dose IL-2 (HDIL2) is approved for the treatment of metastatic melanoma and renal cell carcinoma, but its use is limited in part by toxicity related to the development of vascular leak syndrome (VLS). Therefore, an understanding of the mechanisms that underlie the initiation and progression of HDIL2-induced increases in endothelial cell (EC) permeability leading to VLS are of clinical importance., Methods: We established a novel ex vivo approach utilizing primary human pulmonary microvascular ECs to evaluate EC barrier dysfunction in response to IL-2., Results: Complementary in vitro studies using exogenous IL-2 and ex vivo studies using serum from patients treated with IL-2 demonstrate that HDIL2 induces VLS through CD144 (vascular endothelial (VE)-cadherin) redistribution., Conclusions: These findings provide new insight into how IL-2 induces VLS and identifies VE-cadherin as a potential target for preventing IL-2-related VLS.
- Published
- 2014
- Full Text
- View/download PDF
42. Costimulation via the tumor-necrosis factor receptor superfamily couples TCR signal strength to the thymic differentiation of regulatory T cells.
- Author
-
Mahmud SA, Manlove LS, Schmitz HM, Xing Y, Wang Y, Owen DL, Schenkel JM, Boomer JS, Green JM, Yagita H, Chi H, Hogquist KA, and Farrar MA
- Subjects
- Animals, CD28 Antigens genetics, CD28 Antigens metabolism, Cell Differentiation genetics, Cells, Cultured, Glucocorticoid-Induced TNFR-Related Protein genetics, Glucocorticoid-Induced TNFR-Related Protein metabolism, MAP Kinase Kinase Kinases genetics, MAP Kinase Kinase Kinases metabolism, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Receptors, OX40 genetics, Receptors, OX40 metabolism, Receptors, Tumor Necrosis Factor, Type II metabolism, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins pharmacology, STAT5 Transcription Factor metabolism, Signal Transduction genetics, Tumor Necrosis Factor Receptor-Associated Peptides and Proteins genetics, Receptor Cross-Talk immunology, Receptors, Antigen, T-Cell agonists, T-Lymphocytes, Regulatory immunology, Thymus Gland immunology, Tumor Necrosis Factor Receptor-Associated Peptides and Proteins metabolism
- Abstract
Regulatory T cells (Treg cells) express members of the tumor-necrosis factor (TNF) receptor superfamily (TNFRSF), but the role of those receptors in the thymic development of Treg cells is undefined. We found here that Treg cell progenitors had high expression of the TNFRSF members GITR, OX40 and TNFR2. Expression of those receptors correlated directly with the signal strength of the T cell antigen receptor (TCR) and required the coreceptor CD28 and the kinase TAK1. The neutralization of ligands that are members of the TNF superfamily (TNFSF) diminished the development of Treg cells. Conversely, TNFRSF agonists enhanced the differentiation of Treg cell progenitors by augmenting responsiveness of the interleukin 2 receptor (IL-2R) and transcription factor STAT5. Costimulation with the ligand of GITR elicited dose-dependent enrichment for cells of lower TCR affinity in the Treg cell repertoire. In vivo, combined inhibition of GITR, OX40 and TNFR2 abrogated the development of Treg cells. Thus, expression of members of the TNFRSF on Treg cell progenitors translated strong TCR signals into molecular parameters that specifically promoted the development of Treg cells and shaped the Treg cell repertoire.
- Published
- 2014
- Full Text
- View/download PDF
43. Cutting edge: control of Mycobacterium tuberculosis infection by a subset of lung parenchyma-homing CD4 T cells.
- Author
-
Sakai S, Kauffman KD, Schenkel JM, McBerry CC, Mayer-Barber KD, Masopust D, and Barber DL
- Subjects
- Adoptive Transfer, Animals, Blood Vessels immunology, Blood Vessels metabolism, CD4-Positive T-Lymphocytes metabolism, CD4-Positive T-Lymphocytes transplantation, CX3C Chemokine Receptor 1, Cell Movement immunology, Flow Cytometry, Host-Pathogen Interactions immunology, Interferon-gamma immunology, Interferon-gamma metabolism, Lectins, C-Type, Leukocyte Common Antigens immunology, Leukocyte Common Antigens metabolism, Lung blood supply, Lung microbiology, Mice, Mice, Congenic, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Fluorescence, Mycobacterium tuberculosis physiology, Receptors, CXCR3 immunology, Receptors, CXCR3 metabolism, Receptors, Chemokine immunology, Receptors, Chemokine metabolism, Receptors, Immunologic immunology, Receptors, Immunologic metabolism, Th1 Cells immunology, Th1 Cells metabolism, Tuberculosis microbiology, CD4-Positive T-Lymphocytes immunology, Lung immunology, Mycobacterium tuberculosis immunology, Tuberculosis immunology
- Abstract
Th1 cells are critical for containment of Mycobacterium tuberculosis infection, but little else is known about the properties of protective CD4 T cell responses. In this study, we show that the pulmonary Th1 response against M. tuberculosis is composed of two populations that are either CXCR3(hi) and localize to lung parenchyma or are CX3CR1(hi)KLRG1(hi) and are retained within lung blood vasculature. M. tuberculosis-specific parenchymal CD4 T cells migrate rapidly back into the lung parenchyma upon adoptive transfer, whereas the intravascular effectors produce the highest levels of IFN-γ in vivo. Importantly, parenchymal T cells displayed greater control of infection compared with the intravascular counterparts upon transfer into susceptible T cell-deficient hosts. Thus, we identified a subset of naturally generated M. tuberculosis-specific CD4 T cells with enhanced protective capacity and showed that control of M. tuberculosis correlates with the ability of CD4 T cells to efficiently enter the lung parenchyma rather than produce high levels of IFN-γ.
- Published
- 2014
- Full Text
- View/download PDF
44. Cutting edge: resident memory CD8 T cells occupy frontline niches in secondary lymphoid organs.
- Author
-
Schenkel JM, Fraser KA, and Masopust D
- Subjects
- Animals, Antigens, CD immunology, Antigens, CD metabolism, Antigens, Differentiation, T-Lymphocyte immunology, Antigens, Differentiation, T-Lymphocyte metabolism, CD8-Positive T-Lymphocytes metabolism, Flow Cytometry, Host-Pathogen Interactions immunology, Interleukin-15 deficiency, Interleukin-15 genetics, Interleukin-15 immunology, L-Selectin immunology, L-Selectin metabolism, Lectins, C-Type immunology, Lectins, C-Type metabolism, Lymph Nodes immunology, Lymph Nodes virology, Lymphocytic Choriomeningitis virology, Lymphocytic choriomeningitis virus physiology, Lymphoid Tissue virology, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Microscopy, Fluorescence, Spleen immunology, Spleen virology, CD8-Positive T-Lymphocytes immunology, Immunologic Memory immunology, Lymphocytic Choriomeningitis immunology, Lymphocytic choriomeningitis virus immunology, Lymphoid Tissue immunology
- Abstract
Resident memory CD8 T cells (TRM) are a nonrecirculating subset positioned in nonlymphoid tissues to provide early responses to reinfection. Although TRM are associated with nonlymphoid tissues, we asked whether they populated secondary lymphoid organs (SLO). We show that a subset of virus-specific memory CD8 T cells in SLO exhibit phenotypic signatures associated with TRM, including CD69 expression. Parabiosis revealed that SLO CD69(+) memory CD8 T cells do not circulate, defining them as TRM. SLO TRM were overrepresented in IL-15-deficient mice, suggesting independent regulation compared with central memory CD8 T cells and effector memory CD8 T cells. These cells were positioned at SLO entry points for peripheral Ags: the splenic marginal zone, red pulp, and lymph node sinuses. Consistent with a potential role in guarding SLO pathogen entry points, SLO TRM did not vacate their position in response to peripheral alarm signals. These data extend the range of tissue resident memory to SLO.
- Published
- 2014
- Full Text
- View/download PDF
45. Identification of a resident T-cell memory core transcriptional signature.
- Author
-
Schenkel JM and Masopust D
- Subjects
- Animals, Antigens, CD immunology, CD8-Positive T-Lymphocytes immunology, Immunologic Memory immunology, Integrin alpha Chains immunology, Signal Transduction immunology, Skin immunology
- Published
- 2014
- Full Text
- View/download PDF
46. Preexisting high frequencies of memory CD8+ T cells favor rapid memory differentiation and preservation of proliferative potential upon boosting.
- Author
-
Fraser KA, Schenkel JM, Jameson SC, Vezys V, and Masopust D
- Subjects
- Animals, Antigens, Viral immunology, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes metabolism, Cell Survival immunology, Flow Cytometry, Immunophenotyping, Mice, Mice, Inbred C57BL, Mitochondria immunology, Mitochondria metabolism, Time Factors, CD8-Positive T-Lymphocytes immunology, Cell Differentiation immunology, Cell Proliferation, Immunization, Secondary methods, Immunologic Memory immunology
- Abstract
Memory CD8+ T cell quantity and quality determine protective efficacy against reinfection. Heterologous prime boost vaccination minimizes contraction of anamnestic effectors and maximizes memory CD8+ T cell quantity but reportedly erodes proliferative potential and protective efficacy. This study exploited heterologous prime boost vaccination to discover parameters regulating effector CD8+ T cell contraction and memory differentiation. When abundant memory T cells were established, boosting induced only 5-8 cell divisions, unusually rapid memory T cell differentiation as measured by phenotype and mitochondrial bioenergetic function, long-lived survival of 50% of effector T cells, and preservation of proliferative potential. Conversely, boosting in situations of low memory CD8+ T cell frequencies induced many cell divisions, increased contraction of effector cells, and caused senescence, low mitochondrial membrane potential, and poorly protective memory. Thus, anamnestic memory T cell differentiation is flexible, and abundant quantity can be achieved while maximizing protective efficacy and preserving proliferative potential., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
47. The integration of T cell migration, differentiation and function.
- Author
-
Masopust D and Schenkel JM
- Subjects
- Animals, Cell Differentiation immunology, Humans, Immunologic Memory immunology, Lymphocyte Activation immunology, Lymphoid Tissue cytology, Lymphoid Tissue immunology, T-Lymphocytes classification, T-Lymphocytes cytology, Terminology as Topic, Cell Movement immunology, T-Lymphocytes immunology
- Abstract
T cells function locally. Accordingly, T cells' recognition of antigen, their subsequent activation and differentiation, and their role in the processes of infection control, tumour eradication, autoimmunity, allergy and alloreactivity are intrinsically coupled with migration. Recent discoveries revise our understanding of the regulation and patterns of T cell trafficking and reveal limitations in current paradigms. Here, we review classic and emerging concepts, highlight the challenge of integrating new observations with existing T cell classification schemes and summarize the heuristic framework provided by viewing T cell differentiation and function first through the prism of migration.
- Published
- 2013
- Full Text
- View/download PDF
48. Sensing and alarm function of resident memory CD8⁺ T cells.
- Author
-
Schenkel JM, Fraser KA, Vezys V, and Masopust D
- Subjects
- Animals, Antigens, Viral immunology, Cell Movement immunology, Cells, Cultured, Female, Genitalia, Female immunology, Host-Pathogen Interactions, Interferon-gamma genetics, Interferon-gamma metabolism, Lymphocyte Activation, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Receptors, Antigen, T-Cell, alpha-beta genetics, Transplantation Chimera, Arenaviridae Infections immunology, CD8-Positive T-Lymphocytes immunology, Immunologic Memory, Lymphocytic choriomeningitis virus immunology, T-Lymphocyte Subsets immunology
- Abstract
CD8(+) T cells eliminate intracellular infections through two contact-dependent effector functions: cytolysis and secretion of antiviral cytokines. Here we identify the following additional function for memory CD8(+) T cells that persist at front-line sites of microbial exposure: to serve as local sensors of previously encountered antigens that precipitate innate-like alarm signals and draw circulating memory CD8(+) T cells into the tissue. When memory CD8(+) T cells residing in the female mouse reproductive tract encountered cognate antigen, they expressed interferon-γ (IFN-γ), potentiated robust local expression of inflammatory chemokines and induced rapid recruitment of circulating memory CD8(+) T cells. Anamnestic responses in front-line tissues are thus an integrated collaboration between front-line and circulating populations of memory CD8(+) T cells, and vaccines should establish both populations to maximize rapid responses.
- Published
- 2013
- Full Text
- View/download PDF
49. Antigen-independent differentiation and maintenance of effector-like resident memory T cells in tissues.
- Author
-
Casey KA, Fraser KA, Schenkel JM, Moran A, Abt MC, Beura LK, Lucas PJ, Artis D, Wherry EJ, Hogquist K, Vezys V, and Masopust D
- Subjects
- Animals, Cell Line, Female, Immunophenotyping, Intestinal Mucosa immunology, Intestinal Mucosa pathology, Intestinal Mucosa virology, Lymphocytic choriomeningitis virus immunology, Lymphocytic choriomeningitis virus pathogenicity, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Organ Specificity immunology, Signal Transduction immunology, T-Lymphocyte Subsets pathology, T-Lymphocyte Subsets virology, Time Factors, Tissue Distribution immunology, Cell Differentiation immunology, Epitopes, T-Lymphocyte physiology, Immunologic Memory immunology, T-Lymphocyte Subsets immunology
- Abstract
Differentiation and maintenance of recirculating effector memory CD8 T cells (T(EM)) depends on prolonged cognate Ag stimulation. Whether similar pathways of differentiation exist for recently identified tissue-resident effector memory T cells (T(RM)), which contribute to rapid local protection upon pathogen re-exposure, is unknown. Memory CD8αβ(+) T cells within small intestine epithelium are well-characterized examples of T(RM), and they maintain a long-lived effector-like phenotype that is highly suggestive of persistent Ag stimulation. This study sought to define the sources and requirements for prolonged Ag stimulation in programming this differentiation state, including local stimulation via cognate or cross-reactive Ags derived from pathogens, microbial flora, or dietary proteins. Contrary to expectations, we found that prolonged cognate Ag stimulation was dispensable for intestinal T(RM) ontogeny. In fact, chronic antigenic stimulation skewed differentiation away from the canonical intestinal T cell phenotype. Resident memory signatures, CD69 and CD103, were expressed in many nonlymphoid tissues including intestine, stomach, kidney, reproductive tract, pancreas, brain, heart, and salivary gland and could be driven by cytokines. Moreover, TGF-β-driven CD103 expression was required for T(RM) maintenance within intestinal epithelium in vivo. Thus, induction and maintenance of long-lived effector-like intestinal T(RM) differed from classic models of T(EM) ontogeny and were programmed through a novel location-dependent pathway that was required for the persistence of local immunological memory.
- Published
- 2012
- Full Text
- View/download PDF
50. NKG2D signaling on CD8⁺ T cells represses T-bet and rescues CD4-unhelped CD8⁺ T cell memory recall but not effector responses.
- Author
-
Zloza A, Kohlhapp FJ, Lyons GE, Schenkel JM, Moore TV, Lacek AT, O'Sullivan JA, Varanasi V, Williams JW, Jagoda MC, Bellavance EC, Marzo AL, Thomas PG, Zafirova B, Polić B, Al-Harthi L, Sperling AI, and Guevara-Patiño JA
- Subjects
- Animals, CD4-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes cytology, Cytotoxicity, Immunologic, Disease Models, Animal, Gene Expression, HIV-1 immunology, Humans, Immunity, Cellular, Influenza, Human genetics, Interferon-gamma immunology, Interferon-gamma metabolism, Interleukin-2 immunology, Interleukin-2 metabolism, Mice, Mice, Inbred C57BL, NK Cell Lectin-Like Receptor Subfamily K metabolism, T-Box Domain Proteins metabolism, T-Lymphocytes, Helper-Inducer immunology, Vaccines, DNA immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, HIV Infections immunology, Influenza, Human immunology, NK Cell Lectin-Like Receptor Subfamily K genetics, NK Cell Lectin-Like Receptor Subfamily K immunology
- Abstract
CD4-unhelped CD8(+) T cells are functionally defective T cells primed in the absence of CD4(+) T cell help. Given the co-stimulatory role of natural-killer group 2, member D protein (NKG2D) on CD8(+) T cells, we investigated its ability to rescue these immunologically impotent cells. We demonstrate that augmented co-stimulation through NKG2D during priming paradoxically rescues memory, but not effector, CD8(+) T cell responses. NKG2D-mediated rescue is characterized by reversal of elevated transcription factor T-box expressed in T cells (T-bet) expression and recovery of interleukin-2 and interferon-γ production and cytolytic responses. Rescue is abrogated in CD8(+) T cells lacking NKG2D. Augmented co-stimulation through NKG2D confers a high rate of survival to mice lacking CD4(+) T cells in a CD4-dependent influenza model and rescues HIV-specific CD8(+) T cell responses from CD4-deficient HIV-positive donors. These findings demonstrate that augmented co-stimulation through NKG2D is effective in rescuing CD4-unhelped CD8(+) T cells from their pathophysiological fate and may provide therapeutic benefits.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.