61 results on '"Creg J, Workman"'
Search Results
2. Aplp1 interacts with Lag3 to facilitate transmission of pathologic α-synuclein
- Author
-
Xiaobo Mao, Hao Gu, Donghoon Kim, Yasuyoshi Kimura, Ning Wang, Enquan Xu, Ramhari Kumbhar, Xiaotian Ming, Haibo Wang, Chan Chen, Shengnan Zhang, Chunyu Jia, Yuqing Liu, Hetao Bian, Senthilkumar S. Karuppagounder, Fatih Akkentli, Qi Chen, Longgang Jia, Heehong Hwang, Su Hyun Lee, Xiyu Ke, Michael Chang, Amanda Li, Jun Yang, Cyrus Rastegar, Manjari Sriparna, Preston Ge, Saurav Brahmachari, Sangjune Kim, Shu Zhang, Yasushi Shimoda, Martina Saar, Haiqing Liu, Sin Ho Kweon, Mingyao Ying, Creg J. Workman, Dario A. A. Vignali, Ulrike C. Muller, Cong Liu, Han Seok Ko, Valina L. Dawson, and Ted M. Dawson
- Subjects
Science - Abstract
Abstract Pathologic α-synuclein (α-syn) spreads from cell-to-cell, in part, through binding to the lymphocyte-activation gene 3 (Lag3). Here we report that amyloid β precursor-like protein 1 (Aplp1) interacts with Lag3 that facilitates the binding, internalization, transmission, and toxicity of pathologic α-syn. Deletion of both Aplp1 and Lag3 eliminates the loss of dopaminergic neurons and the accompanying behavioral deficits induced by α-syn preformed fibrils (PFF). Anti-Lag3 prevents the internalization of α-syn PFF by disrupting the interaction of Aplp1 and Lag3, and blocks the neurodegeneration induced by α-syn PFF in vivo. The identification of Aplp1 and the interplay with Lag3 for α-syn PFF induced pathology deepens our insight about molecular mechanisms of cell-to-cell transmission of pathologic α-syn and provides additional targets for therapeutic strategies aimed at preventing neurodegeneration in Parkinson’s disease and related α-synucleinopathies.
- Published
- 2024
- Full Text
- View/download PDF
3. Lymphocyte‐Activation Gene 3 Facilitates Pathological Tau Neuron‐to‐Neuron Transmission
- Author
-
Chan Chen, Ramhari Kumbhar, Hu Wang, Xiuli Yang, Kundlik Gadhave, Cyrus Rastegar, Yasuyoshi Kimura, Adam Behensky, Sumasri Kotha, Grace Kuo, Sruthi Katakam, Deok Jeong, Liang Wang, Anthony Wang, Rong Chen, Shu Zhang, Lingtao Jin, Creg J. Workman, Dario A. A. Vignali, Olga Pletinkova, Hongpeng Jia, Weiyi Peng, David W. Nauen, Philip C. Wong, Javier Redding‐Ochoa, Juan C. Troncoso, Mingyao Ying, Valina L. Dawson, Ted M. Dawson, and Xiaobo Mao
- Subjects
lymphocyte‐activation gene 3 ,cell‐to‐cell transmission ,receptor ,Tau ,Tau preformed fibrils ,Science - Abstract
Abstract The spread of prion‐like protein aggregates is a common driver of pathogenesis in various neurodegenerative diseases, including Alzheimer's disease (AD) and related Tauopathies. Tau pathologies exhibit a clear progressive spreading pattern that correlates with disease severity. Clinical observation combined with complementary experimental studies has shown that Tau preformed fibrils (PFF) are prion‐like seeds that propagate pathology by entering cells and templating misfolding and aggregation of endogenous Tau. While several cell surface receptors of Tau are known, they are not specific to the fibrillar form of Tau. Moreover, the underlying cellular mechanisms of Tau PFF spreading remain poorly understood. Here, it is shown that the lymphocyte‐activation gene 3 (Lag3) is a cell surface receptor that binds to PFF but not the monomer of Tau. Deletion of Lag3 or inhibition of Lag3 in primary cortical neurons significantly reduces the internalization of Tau PFF and subsequent Tau propagation and neuron‐to‐neuron transmission. Propagation of Tau pathology and behavioral deficits induced by injection of Tau PFF in the hippocampus and overlying cortex are attenuated in mice lacking Lag3 selectively in neurons. These results identify neuronal Lag3 as a receptor of pathologic Tau in the brain,and for AD and related Tauopathies, a therapeutic target.
- Published
- 2024
- Full Text
- View/download PDF
4. Regulatory T Cells: Barriers of Immune Infiltration Into the Tumor Microenvironment
- Author
-
Ellen N. Scott, Angela M. Gocher, Creg J. Workman, and Dario A. A. Vignali
- Subjects
regulatory T cells (Treg) ,immune infiltration ,tumor microenvironment ,cancer ,vasculature ,stroma ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Regulatory T cells (Tregs) are key immunosuppressive cells that promote tumor growth by hindering the effector immune response. Tregs utilize multiple suppressive mechanisms to inhibit pro-inflammatory responses within the tumor microenvironment (TME) by inhibition of effector function and immune cell migration, secretion of inhibitory cytokines, metabolic disruption and promotion of metastasis. In turn, Tregs are being targeted in the clinic either alone or in combination with other immunotherapies, in efforts to overcome the immunosuppressive TME and increase anti-tumor effects. However, it is now appreciated that Tregs not only suppress cells intratumorally via direct engagement, but also serve as key interactors in the peritumor, stroma, vasculature and lymphatics to limit anti-tumor immune responses prior to tumor infiltration. We will review the suppressive mechanisms that Tregs utilize to alter immune and non-immune cells outside and within the TME and discuss how these mechanisms collectively allow Tregs to create and promote a physical and biological barrier, resulting in an immune-excluded or limited tumor microenvironment.
- Published
- 2021
- Full Text
- View/download PDF
5. IFNγ-induction of TH1-like regulatory T cells controls antiviral responses
- Author
-
Angela M. Gocher-Demske, Jian Cui, Andrea L. Szymczak-Workman, Kate M. Vignali, Julianna N. Latini, Gwen P. Pieklo, Jesse C. Kimball, Lyndsay Avery, Ellyse M. Cipolla, Brydie R. Huckestein, Lee Hedden, Marlies Meisel, John F. Alcorn, Lawrence P. Kane, Creg J. Workman, and Dario A. A. Vignali
- Subjects
Immunology ,Immunology and Allergy ,Article - Abstract
Regulatory T (T(reg)) cells are an immunosuppressive population that are required to maintain peripheral tolerance and prevent tissue damage from immunopathology, via anti-inflammatory cytokines, inhibitor receptors and metabolic disruption. Here we show that T(reg) cells acquire an effector-like state, yet remain stable and functional, when exposed to interferon gamma (IFNγ) during infection with lymphocytic choriomeningitis and influenza A virus. T(reg) cell-restricted deletion of the IFNγ receptor (encoded by Ifngr1), but not the interleukin 12 (IL12) receptor (encoded by Il12rb2), prevented T(H)1-like polarization (decreased expression of T-bet, CXC motif chemokine receptor 3 and IFNγ) and promoted T(H)2-like polarization (increased expression of GATA-3, CCR4 and IL4). T(H)1-like T(reg) cells limited CD8(+) T cell effector function, proliferation and memory formation during acute and chronic infection. These findings provide fundamental insights into how T(reg) cells sense inflammatory cues from the environment (such as IFNγ) during viral infection to provide guidance to the effector immune response. This regulatory circuit prevents prolonged immunoinflammatory responses and shapes the quality and quantity of the memory T cell response.
- Published
- 2023
6. Treg-Cell-Derived IL-35-Coated Extracellular Vesicles Promote Infectious Tolerance
- Author
-
Jeremy A. Sullivan, Yusuke Tomita, Ewa Jankowska-Gan, Diego A. Lema, Matt P. Arvedson, Ashita Nair, William Bracamonte-Baran, Ying Zhou, Kristy K. Meyer, Weixiong Zhong, Deepali V. Sawant, Andrea L. Szymczak-Workman, Qianxia Zhang, Creg J. Workman, Seungpyo Hong, Dario A.A. Vignali, and William J. Burlingham
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Interleukin-35 (IL-35) is an immunosuppressive cytokine composed of Epstein-Barr-virus-induced protein 3 (Ebi3) and IL-12α chain (p35) subunits, yet the forms that IL-35 assume and its role in peripheral tolerance remain elusive. We induce CBA-specific, IL-35-producing T regulatory (Treg) cells in TregEbi3WT C57BL/6 reporter mice and identify IL-35 producers by expression of Ebi3TdTom gene reporter plus Ebi3 and p35 proteins. Curiously, both subunits of IL-35 are displayed on the surface of tolerogen-specific Foxp3+ and Foxp3neg (iTr35) T cells. Furthermore, IL-35 producers, although rare, secrete Ebi3 and p35 on extracellular vesicles (EVs) targeting a 25- to 100-fold higher number of T and B lymphocytes, causing them to acquire surface IL-35. This surface IL-35 is absent when EV production is inhibited or if Ebi3 is genetically deleted in Treg cells. The unique ability of EVs to coat bystander lymphocytes with IL-35, promoting exhaustion in, and secondary suppression by, non-Treg cells identifies a novel mechanism of infectious tolerance. : Sullivan et al. show that while many factors and cytokines contribute to primary immunosuppression, EV-associated IL-35 uniquely promotes “infectious” tolerance not only by inducing IL-35 production in non-Treg cells but also by causing an immunosuppressive phenotype in EV-acquiring T and B cells, leading to secondary suppression of immune responses. Keywords: IL-35, extracellular vesicles, cytokines, tolerance, Treg, tetraspanin, Ebi3, p35, CD81
- Published
- 2020
- Full Text
- View/download PDF
7. Neuronally expressed PDL1, not PD1, suppresses acute nociception
- Author
-
Kimberly A. Meerschaert, Brian S. Edwards, Ariel Y. Epouhe, Bahiyyah Jefferson, Robert Friedman, Olivia L. Babyok, Jamie K. Moy, Faith Kehinde, Chang Liu, Creg J. Workman, Dario A.A. Vignali, Kathryn M. Albers, H. Richard Koerber, Michael S. Gold, Brian M. Davis, Nicole N. Scheff, and Jami L. Saloman
- Subjects
Nociception ,Mice ,Behavioral Neuroscience ,Endocrine and Autonomic Systems ,Immunology ,Animals ,Calcium ,RNA, Messenger ,Capsaicin ,B7-H1 Antigen - Abstract
PDL1 is a protein that induces immunosuppression by binding to PD1 expressed on immune cells. In line with historical studies, we found that membrane-bound PD1 expression was largely restricted to immune cells; PD1 was not detectable at either the mRNA or protein level in peripheral neurons using single neuron qPCR, immunolabeling and flow cytometry. However, we observed widespread expression of PDL1 in both sensory and sympathetic neurons that could have important implications for patients receiving immunotherapies targeting this pathway that include unexpected autonomic and sensory related effects. While signaling pathways downstream of PD1 are well established, little to no information is available regarding the intracellular signaling downstream of membrane-bound PDL1 (also known as reverse signaling). Here, we administered soluble PD1 to engage neuronally expressed PDL1 and found that PD1 significantly reduced nocifensive behaviors evoked by algogenic capsaicin. We used calcium imaging to examine the underlying neural mechanism of this reduction and found that exogenous PD1 diminished TRPV1-dependent calcium transients in dissociated sensory neurons. Furthermore, we observed a reduction in membrane expression of TRPV1 following administration of PD1. Exogenous PD1 had no effect on pain-related behaviors in sensory neuron specific PDL1 knockout mice. These data indicate that neuronal PDL1 activation is sufficient to modulate sensitivity to noxious stimuli and as such, may be an important homeostatic mechanism for regulating acute nociception.
- Published
- 2022
8. Pathological Tau transmission initiated by binding lymphocyte-activation gene 3
- Author
-
Chan Chen, Ramhari Kumbhar, Hu Wang, Xiuli Yang, Kundlik Gadhave, Cyrus Rastegar, Yasuyoshi Kimura, Adam Behensky, Sruthi Katakam, Deok Jeong, Liang Wang, Anthony Wang, Rong Chen, Shu Zhang, Lingtao Jin, Creg J. Workman, Dario A.A. Vignali, Olga Pletinkova, David W. Nauen, Philip C. Wong, Juan C. Troncoso, Mingyao Ying, Valina L. Dawson, Ted M. Dawson, and Xiaobo Mao
- Subjects
Article - Abstract
The spread of prion-like protein aggregates is believed to be a common driver of pathogenesis in many neurodegenerative diseases. Accumulated tangles of filamentous Tau protein are considered pathogenic lesions of Alzheimer’s disease (AD) and related Tauopathies, including progressive supranuclear palsy, and corticobasal degeneration. Tau pathologies in these illnesses exhibits a clear progressive and hierarchical spreading pattern that correlates with disease severity1, 2. Clinical observation combined with complementary experimental studies3, 4have shown that Tau preformed fibrils (PFF) are prion-like seeds that propagate pathology by entering cells and templating misfolding and aggregation of endogenous Tau. While several receptors of Tau are known, they are not specific to the fibrillar form of Tau. Moreover, the underlying cellular mechanisms of Tau PFF spreading remains poorly understood. Here, we show that the lymphocyte-activation gene 3 (Lag3) is a cell surface receptor that binds to PFF, but not monomer, of Tau. Deletion ofLag3or inhibition of Lag3 in primary cortical neurons significantly reduces the internalization of Tau PFF and subsequent Tau propagation and neuron-to-neuron transmission. Propagation of Tau pathology and behavioral deficits induced by injection of Tau PFF in the hippocampus and overlying cortex are attenuated in mice lackingLag3selectively in neurons. Our results identify neuronal Lag3 as a receptor of pathologic Tau in the brain, and for AD and related Tauopathies a therapeutic target.One Sentence SummaryLag3 is a neuronal receptor specific for Tau PFF, and is required for uptake, propagation and transmission of Tau pathology.
- Published
- 2023
9. Supplementary Data from Systemic Immune Dysfunction in Cancer Patients Driven by IL6 Induction of LAG3 in Peripheral CD8+ T Cells
- Author
-
Dario A.A. Vignali, Tullia C. Bruno, Robert L. Ferris, Evan J. Lipson, John M. Kirkwood, James G. Herman, Simon C. Watkins, Daniel P. Normolle, Dhivyaa Rajasundaram, Rebekah Dadey, Michael Calderon, Sonali Joyce, Maria Velez, Lauren Oliveri, Sheryl Kunning, Creg J. Workman, Caleb Lampenfeld, Anthony R. Cillo, and Ashwin Somasundaram
- Abstract
Supplementary Data from Systemic Immune Dysfunction in Cancer Patients Driven by IL6 Induction of LAG3 in Peripheral CD8+ T Cells
- Published
- 2023
10. Data from Systemic Immune Dysfunction in Cancer Patients Driven by IL6 Induction of LAG3 in Peripheral CD8+ T Cells
- Author
-
Dario A.A. Vignali, Tullia C. Bruno, Robert L. Ferris, Evan J. Lipson, John M. Kirkwood, James G. Herman, Simon C. Watkins, Daniel P. Normolle, Dhivyaa Rajasundaram, Rebekah Dadey, Michael Calderon, Sonali Joyce, Maria Velez, Lauren Oliveri, Sheryl Kunning, Creg J. Workman, Caleb Lampenfeld, Anthony R. Cillo, and Ashwin Somasundaram
- Abstract
Many cancer patients do not develop a durable response to the current standard-of-care immunotherapies, despite substantial advances in targeting immune inhibitory receptors. A potential compounding issue, which may serve as an unappreciated, dominant resistance mechanism, is an inherent systemic immune dysfunction that is often associated with advanced cancer. Minimal response to inhibitory receptor (IR) blockade therapy and increased disease burden have been associated with peripheral CD8+ T-cell dysfunction, characterized by suboptimal T-cell proliferation and chronic expression of IRs (e.g., PD1 and LAG3). Here, we demonstrated that approximately a third of cancer patients analyzed in this study have peripheral CD8+ T cells that expressed robust intracellular LAG3 (LAG3IC), but not surface LAG3 (LAG3SUR) due to a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) cleavage. This is associated with poor disease prognosis and decreased CD8+ T-cell function, which could be partially reversed by anti-LAG3. Systemic immune dysfunction was restricted to CD8+ T cells, including, in some cases, a high percentage of peripheral naïve CD8+ T cells, and was driven by the cytokine IL6 via STAT3. These data suggest that additional studies are warranted to determine if the combination of increased LAG3IC in peripheral CD8+ T cells and elevated systemic IL6 can serve as predictive biomarkers and identify which cancer patients may benefit from LAG3 blockade.
- Published
- 2023
11. Supplementary Tables 1-2, Figures 1-11 from Immune Inhibitory Molecules LAG-3 and PD-1 Synergistically Regulate T-cell Function to Promote Tumoral Immune Escape
- Author
-
Dario A.A. Vignali, Charles G. Drake, Alan J. Korman, Drew M. Pardoll, Creg J. Workman, Paul J. Utz, Alcides Chaux, Matthew P. Smeltzer, George Netto, Joseph F. Grosso, Stephanie Tangsombatvisit, Chih Long Liu, Peter Vogel, David M. Gravano, Matthew L. Bettini, Christopher J. Nirschl, Mark Selby, Jaishree Bankoti, Monica V. Goldberg, Meghan E. Turnis, and Seng-Ryong Woo
- Abstract
PDF file - 1.1MB
- Published
- 2023
12. Autoreactive CD8+ T cells are restrained by an exhaustion-like program that is maintained by LAG3
- Author
-
Stephanie Grebinoski, Qianxia Zhang, Anthony R. Cillo, Sasikanth Manne, Hanxi Xiao, Erin A. Brunazzi, Tracy Tabib, Carly Cardello, Christine G. Lian, George F. Murphy, Robert Lafyatis, E. John Wherry, Jishnu Das, Creg J. Workman, and Dario A. A. Vignali
- Subjects
Immunology ,Immunology and Allergy - Published
- 2022
13. Systemic Immune Dysfunction in Cancer Patients Driven by IL6 Induction of LAG3 in Peripheral CD8+ T Cells
- Author
-
Ashwin Somasundaram, Anthony R. Cillo, Caleb Lampenfeld, Creg J. Workman, Sheryl Kunning, Lauren Oliveri, Maria Velez, Sonali Joyce, Michael Calderon, Rebekah Dadey, Dhivyaa Rajasundaram, Daniel P. Normolle, Simon C. Watkins, James G. Herman, John M. Kirkwood, Evan J. Lipson, Robert L. Ferris, Tullia C. Bruno, and Dario A.A. Vignali
- Subjects
Cancer Research ,Antigens, CD ,Interleukin-6 ,Neoplasms ,Immunology ,Humans ,Immunotherapy ,CD8-Positive T-Lymphocytes ,Receptors, Immunologic ,Lymphocyte Activation Gene 3 Protein ,Article - Abstract
Many cancer patients do not develop a durable response to the current standard-of-care immunotherapies, despite substantial advances in targeting immune inhibitory receptors. A potential compounding issue, which may serve as an unappreciated, dominant resistance mechanism, is an inherent systemic immune dysfunction that is often associated with advanced cancer. Minimal response to inhibitory receptor (IR) blockade therapy and increased disease burden have been associated with peripheral CD8+ T-cell dysfunction, characterized by suboptimal T-cell proliferation and chronic expression of IRs (e.g., PD1 and LAG3). Here, we demonstrated that approximately a third of cancer patients analyzed in this study have peripheral CD8+ T cells that expressed robust intracellular LAG3 (LAG3IC), but not surface LAG3 (LAG3SUR) due to a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) cleavage. This is associated with poor disease prognosis and decreased CD8+ T-cell function, which could be partially reversed by anti-LAG3. Systemic immune dysfunction was restricted to CD8+ T cells, including, in some cases, a high percentage of peripheral naïve CD8+ T cells, and was driven by the cytokine IL6 via STAT3. These data suggest that additional studies are warranted to determine if the combination of increased LAG3IC in peripheral CD8+ T cells and elevated systemic IL6 can serve as predictive biomarkers and identify which cancer patients may benefit from LAG3 blockade.
- Published
- 2022
14. LAG3 associates with TCR–CD3 complexes and suppresses signaling by driving co-receptor–Lck dissociation
- Author
-
Clifford Guy, Diana M. Mitrea, Po-Chien Chou, Jamshid Temirov, Kate M. Vignali, Xueyan Liu, Hui Zhang, Richard Kriwacki, Marcel P. Bruchez, Simon C. Watkins, Creg J. Workman, and Dario A. A. Vignali
- Subjects
Immunology ,Immunology and Allergy - Published
- 2022
15. COVID-19 versus Non–COVID-19 Acute Respiratory Distress Syndrome: Comparison of Demographics, Physiologic Parameters, Inflammatory Biomarkers, and Clinical Outcomes
- Author
-
Georgios D Kitsios, Dario A. A. Vignali, Feng Shan, Callie Drohan, N. Bensen, Haopu Yang, Rebecca S. DeSensi, William Bain, Barbara Methé, Yingze Zhang, Bryan J. McVerry, Tullia C. Bruno, Caleb Lampenfeld, Nameer Al-Yousif, Alison Morris, Tomeka Suber, Carly Cardello, Faraaz Ali Shah, Janet S. Lee, Brian R. Rosborough, Prabir Ray, Anthony R. Cillo, Creg J. Workman, Ashwin Somasundaram, Caitlin Schaefer, and Anuradha Ray
- Subjects
Pulmonary and Respiratory Medicine ,ARDS ,medicine.medical_specialty ,Critical Care ,medicine.medical_treatment ,Lung injury ,Pulmonary compliance ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,pneumonia ,030212 general & internal medicine ,Prospective cohort study ,Original Research ,Mechanical ventilation ,business.industry ,SARS-CoV-2 ,COVID-19 ,acute respiratory distress syndrome ,medicine.disease ,Pneumonia ,030228 respiratory system ,business ,Respiratory minute volume ,Cohort study - Abstract
Rationale: There is an urgent need for improved understanding of the mechanisms and clinical characteristics of acute respiratory distress syndrome (ARDS) due to coronavirus disease (COVID-19). Objectives: To compare key demographic and physiologic parameters, biomarkers, and clinical outcomes of COVID-19 ARDS and ARDS secondary to direct lung injury from other etiologies of pneumonia. Methods: We enrolled 27 patients with COVID-19 ARDS in a prospective, observational cohort study and compared them with a historical, pre–COVID-19 cohort of patients with viral ARDS (n = 14), bacterial ARDS (n = 21), and ARDS due to culture-negative pneumonia (n = 30). We recorded clinical demographics; measured respiratory mechanical parameters; collected serial peripheral blood specimens for measurement of plasma interleukin (IL)-6, IL-8, and IL-10; and followed patients prospectively for patient-centered outcomes. We conducted between-group comparisons with nonparametric tests and analyzed time-to-event outcomes with Kaplan-Meier and Cox proportional hazards models. Results: Patients with COVID-19 ARDS had higher body mass index and were more likely to be Black, or residents of skilled nursing facilities, compared with those with non–COVID-19 ARDS (P < 0.05). Patients with COVID-19 had lower delivered minute ventilation compared with bacterial and culture-negative ARDS (post hoc P < 0.01) but not compared with viral ARDS. We found no differences in static compliance, hypoxemic indices, or carbon dioxide clearance between groups. Patients with COVID-19 had lower IL-6 levels compared with bacterial and culture-negative ARDS at early time points after intubation but no differences in IL-6 levels compared with viral ARDS. Patients with COVID-19 had longer duration of mechanical ventilation but similar 60-day mortality in both unadjusted and adjusted analyses. Conclusions: COVID-19 ARDS bears several similarities to viral ARDS but demonstrates lower minute ventilation and lower systemic levels of IL-6 compared with bacterial and culture-negative ARDS. COVID-19 ARDS was associated with longer dependence on mechanical ventilation compared with non–COVID-19 ARDS. Such detectable differences of COVID-19 do not merit deviation from evidence-based management of ARDS but suggest priorities for clinical research to better characterize and treat this new clinical entity.
- Published
- 2021
16. Regulatory T Cell–Derived TRAIL Is Not Required for Peripheral Tolerance
- Author
-
Stephanie Grebinoski, Erin A. Brunazzi, Rebekah E. Dadey, Qianxia Zhang, Dario A. A. Vignali, Amanda R. Burton, and Creg J. Workman
- Subjects
Male ,Programmed cell death ,Encephalomyelitis, Autoimmune, Experimental ,Regulatory T cell ,Immunology ,Apoptosis ,Nod ,Lymphocyte Activation ,T-Lymphocytes, Regulatory ,Article ,Myelin oligodendrocyte glycoprotein ,TNF-Related Apoptosis-Inducing Ligand ,Mice ,Immune system ,Mice, Inbred NOD ,Neoplasms ,Conditional gene knockout ,medicine ,Animals ,Immunology and Allergy ,Mice, Knockout ,Mice, Inbred BALB C ,biology ,Peripheral Tolerance ,Experimental autoimmune encephalomyelitis ,Peripheral tolerance ,General Medicine ,Flow Cytometry ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,biology.protein ,Cancer research ,Female ,Myelin-Oligodendrocyte Glycoprotein - Abstract
TRAIL (Tnfsf10/TRAIL/CD253/Apo2L) is an important immune molecule that mediates apoptosis. TRAIL can play key roles in regulating cell death in the tumor and autoimmune microenvironments. However, dissecting TRAIL function remains difficult because of the lack of optimal models. We have now generated a conditional knockout (Tnfsf10L/L) for cell type–specific analysis of TRAIL function on C57BL/6, BALB/c, and NOD backgrounds. Previous studies have suggested a role for TRAIL in regulatory T cell (Treg)–mediated suppression. We generated mice with a Treg-restricted Tnfsf10 deletion and surprisingly found no impact on tumor growth in C57BL/6 and BALB/c tumor models. Furthermore, we found no difference in the suppressive capacity of Tnfsf10-deficient Tregs and no change in function or proliferation of T cells in tumors. We also assessed the role of TRAIL on Tregs in two autoimmune mouse models: the NOD mouse model of autoimmune diabetes and the myelin oligodendrocyte glycoprotein (MOG) C57BL/6 model of experimental autoimmune encephalomyelitis. We found that deletion of Tnfsf10 on Tregs had no effect on disease progression in either model. We conclude that Tregs do not appear to be dependent on TRAIL exclusively as a mechanism of suppression in both the tumor and autoimmune microenvironments, although it remains possible that TRAIL may contribute in combination with other mechanisms and/or in different disease settings. Our Tnfsf10 conditional knockout mouse should prove to be a useful tool for the dissection of TRAIL function on different cell populations in multiple mouse models of human disease.
- Published
- 2021
17. Epitope Mapping of Therapeutic Antibodies Targeting Human LAG3
- Author
-
Pragati Agnihotri, Arjun K. Mishra, Priyanka Agarwal, Kate M. Vignali, Creg J. Workman, Dario A. A. Vignali, and Roy A. Mariuzza
- Subjects
Epitopes ,Mice ,Antigens, CD ,T-Lymphocytes ,Immunology ,Immunology and Allergy ,Animals ,Antibodies, Monoclonal ,Humans ,Lymphocyte Activation Gene 3 Protein ,Epitope Mapping ,Single-Chain Antibodies - Abstract
Lymphocyte activation gene 3 protein (LAG3; CD223) is an inhibitory receptor that is highly upregulated on exhausted T cells in tumors and chronic viral infection. Consequently, LAG3 is now a major immunotherapeutic target for the treatment of cancer, and many mAbs against human (h) LAG3 (hLAG3) have been generated to block its inhibitory activity. However, little or no information is available on the epitopes they recognize. We selected a panel of seven therapeutic mAbs from the patent literature for detailed characterization. These mAbs were expressed as Fab or single-chain variable fragments and shown to bind hLAG3 with nanomolar affinities, as measured by biolayer interferometry. Using competitive binding assays, we found that the seven mAbs recognize four distinct epitopes on hLAG3. To localize the epitopes, we carried out epitope mapping using chimeras between hLAG3 and mouse LAG3. All seven mAbs are directed against the first Ig-like domain (D1) of hLAG3, despite their different origins. Three mAbs almost exclusively target a unique 30-residue loop of D1 that forms at least part of the putative binding site for MHC class II, whereas four mainly recognize D1 determinants outside this loop. However, because all the mAbs block binding of hLAG3 to MHC class II, each of the epitopes they recognize must at least partially overlap the MHC class II binding site.
- Published
- 2022
18. Autoreactive CD8
- Author
-
Stephanie, Grebinoski, Qianxia, Zhang, Anthony R, Cillo, Sasikanth, Manne, Hanxi, Xiao, Erin A, Brunazzi, Tracy, Tabib, Carly, Cardello, Christine G, Lian, George F, Murphy, Robert, Lafyatis, E John, Wherry, Jishnu, Das, Creg J, Workman, and Dario A A, Vignali
- Subjects
Phenotype ,Neoplasms ,Humans ,Autoimmunity ,CD8-Positive T-Lymphocytes - Abstract
Impaired chronic viral and tumor clearance has been attributed to CD8
- Published
- 2022
19. Therapeutic targeting of regulatory T cells in cancer
- Author
-
Feng Shan, Ashwin Somasundaram, Tullia C. Bruno, Creg J. Workman, and Dario A.A. Vignali
- Subjects
Cancer Research ,Oncology ,Neoplasms ,Tumor Microenvironment ,Humans ,Autoimmunity ,Immunotherapy ,T-Lymphocytes, Regulatory - Abstract
The success of immunotherapy in oncology underscores the vital role of the immune system in cancer development. Regulatory T cells (Tregs) maintain a fine balance between autoimmunity and immune suppression. They have multiple roles in the tumor microenvironment (TME) but act particularly in suppressing T cell activation. This review focuses on the detrimental and sometimes beneficial roles of Tregs in tumors, our current understanding of recruitment and stabilization of Tregs within the TME, and current Treg-targeted therapeutics. Research identifying subpopulations of Tregs and their respective functions and interactions within the complex networks of the TME will be crucial to develop the next generation of immunotherapies. Through these advances, Treg-targeted immunotherapy could have important implications for the future of oncology.
- Published
- 2022
20. Neuropilin-1 is a T cell memory checkpoint limiting long-term antitumor immunity
- Author
-
Robert L. Ferris, E. John Wherry, Andrea L. Szymczak-Workman, Ellen N. Scott, Evan J. Lipson, Creg J. Workman, Sasikanth Manne, Dario A. A. Vignali, Ashwin Somasundaram, Angela M. Gocher, Daniel P. Normolle, Tullia C. Bruno, Kate M. Vignali, and Chang Liu
- Subjects
0301 basic medicine ,T cell ,medicine.medical_treatment ,Immunology ,T-cell receptor ,Immunotherapy ,Biology ,Immune checkpoint ,Immune tolerance ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,Memory cell ,medicine ,Cancer research ,Immunology and Allergy ,CD8 ,030215 immunology - Abstract
Robust CD8+ T cell memory is essential for long-term protective immunity but is often compromised in cancer, where T cell exhaustion leads to loss of memory precursors. Immunotherapy via checkpoint blockade may not effectively reverse this defect, potentially underlying disease relapse. Here we report that mice with a CD8+ T cell–restricted neuropilin-1 (NRP1) deletion exhibited substantially enhanced protection from tumor rechallenge and sensitivity to anti-PD1 immunotherapy, despite unchanged primary tumor growth. Mechanistically, NRP1 cell-intrinsically limited the self-renewal of the CD44+PD1+TCF1+TIM3− progenitor exhausted T cells, which was associated with their reduced ability to induce c-Jun/AP-1 expression on T cell receptor restimulation, a mechanism that may contribute to terminal T cell exhaustion at the cost of memory differentiation in wild-type tumor-bearing hosts. These data indicate that blockade of NRP1, a unique ‘immune memory checkpoint’, may promote the development of long-lived tumor-specific Tmem that are essential for durable antitumor immunity. T cell exhaustion limits antitumor immune responses. Vignali and colleagues identify neuropilin-1 as a novel immune checkpoint that cell-intrinsically operates to limit memory cell formation and can be targeted to enhance antitumor responses.
- Published
- 2020
21. Molecular Pathways and Mechanisms of LAG3 in Cancer Therapy
- Author
-
Lawrence P. Andrews, Anthony R. Cillo, Lilit Karapetyan, John M. Kirkwood, Creg J. Workman, and Dario A.A. Vignali
- Subjects
Cancer Research ,Oncology ,Clinical Trials, Phase III as Topic ,Antigens, CD ,Programmed Cell Death 1 Receptor ,Humans ,Immunotherapy ,CD8-Positive T-Lymphocytes ,Melanoma ,Article - Abstract
Immunotherapy targeting coinhibitory receptors has been highly successful in treating a wide variety of malignancies; however, only a subset of patients exhibits durable responses. The first FDA-approved immunotherapeutics targeting coinhibitory receptors PD1 and CTLA4, alone or in combination, significantly improved survival but were also accompanied by substantial toxicity in combination. The third FDA-approved immune checkpoint inhibitor targets LAG3, a coinhibitory receptor expressed on activated CD4+ and CD8+ T cells, especially in settings of long-term antigenic stimulation, such as chronic viral infection or cancer. Mechanistically, LAG3 expression limits both the expansion of activated T cells and the size of the memory pool, suggesting that LAG3 may be a promising target for immunotherapy. Importantly, the mechanism(s) by which LAG3 contributes to CD8+ T-cell exhaustion may be distinct from those governed by PD1, indicating that the combination of anti-LAG3 and anti-PD1 may synergistically enhance antitumor immunity. Clinical studies evaluating the role of anti-LAG3 in combination with anti-PD1 are underway, and recent phase III trial results in metastatic melanoma demonstrate both the efficacy and safety of this combination. Further ongoing clinical trials are evaluating this combination across multiple tumor types and the adjuvant setting, with accompanying translational and biomarker-focused studies designed to elucidate the molecular pathways that lead to improved antitumor T-cell responses following dual blockade of PD1 and LAG3. Overall, LAG3 plays an important role in limiting T-cell activation and has now become part of the repertoire of combinatorial immunotherapeutics available for the treatment of metastatic melanoma.
- Published
- 2022
22. Antibodies targeting conserved non-canonical antigens and endemic coronaviruses associate with favorable outcomes in severe COVID-19
- Author
-
Sai Preetham Peddireddy, Syed A. Rahman, Anthony R. Cillo, Godhev Manakkat Vijay, Ashwin Somasundaram, Creg J. Workman, William Bain, Bryan J. McVerry, Barbara Methe, Janet S. Lee, Prabir Ray, Anuradha Ray, Tullia C. Bruno, Dario A. A. Vignali, Georgios D. Kitsios, Alison Morris, Harinder Singh, Aniruddh Sarkar, and Jishnu Das
- Subjects
SARS-CoV-2 ,Spike Glycoprotein, Coronavirus ,COVID-19 ,Humans ,Antibodies, Viral ,Pandemics ,General Biochemistry, Genetics and Molecular Biology - Abstract
While there have been extensive analyses characterizing cellular and humoral responses across the severity spectrum in COVID-19, predictors of outcomes within severe COVID-19 remain to be comprehensively elucidated. Recently, we identified divergent monocyte states as predictors of outcomes within severe COVID-19, but corresponding humoral profiles of risk have not been delineated. Furthermore, the nature of antibodies (Abs) directed against viral antigens beyond the spike protein or endemic coronavirus antigens and their associations with disease severity and outcomes remain poorly defined. We performed deep molecular profiling of Abs directed against a wide range of antigenic specificities in severe COVID-19 patients admitted to the ICU. The profiles consisted of canonical (S, RBD, N) and non-canonical (orf3a, orf8, nsp3, nps13 and M) antigenic specificities. Notably, multivariate machine learning (ML) models, generated using profiles of Abs directed against canonical or non-canonical antigens, were equally discriminative of recovery and mortality COVID-19 outcomes. In both ML models, survivors were associated with increased virus-specific IgA and IgG3 antibodies and with higher antigen-specific antibody galactosylation. Intriguingly, pre-pandemic healthy controls had cross-reactive Abs directed against nsp13 which is a conserved protein in other alpha and beta coronaviruses. Notably, higher levels of nsp13-specific IgA antibodies were associated with recovery in severe COVID-19. In keeping with these findings, a model built on Ab profiles for endemic coronavirus antigens was also predictive of COVID-19 outcome bifurcation, with higher levels of IgA and IgG3 antibodies against OC43 S and NL63 S being associated with survival. Our results suggest the importance of Abs targeting non-canonical SARS-CoV-2 antigens as well as those directed against endemic coronaviruses in favorable outcomes of severe COVID-19.
- Published
- 2021
23. Intractable Coronavirus Disease 2019 (COVID-19) and Prolonged Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Replication in a Chimeric Antigen Receptor-Modified T-Cell Therapy Recipient: A Case Study
- Author
-
Matthew K Hensley, Jana L. Jacobs, Bernard J.C. Macatangay, Rahil Sethi, Ashwin Somasundaram, Feng Shan, Ghady Haidar, Brittany Staines, Anthony R. Cillo, Janet S. Lee, Linda J. Rennick, Amy Heaps, Anuradha Ray, Michele D. Sobolewski, William Bain, Barbara Methé, Tullia C. Bruno, Prabir Ray, Dario A. A. Vignali, John W. Mellors, William E. Schwarzmann, Georgios D Kitsios, W. Paul Duprex, Urvi M. Parikh, Creg J. Workman, Carly Cardello, Cynthia Klamar-Blain, Mounzer Agha, Sham Nambulli, Kevin D. McCormick, Mark S. Ladinsky, Pamela J. Bjorkman, and Alison Morris
- Subjects
0301 basic medicine ,Microbiology (medical) ,T cell ,viruses ,Cell- and Tissue-Based Therapy ,Virus Replication ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Immunity ,medicine ,Humans ,030212 general & internal medicine ,Coronavirus ,Receptors, Chimeric Antigen ,SARS-CoV-2 ,Transmission (medicine) ,business.industry ,Brief Report ,SARS-CoV-2 infectivity ,SARS-CoV-2 intrahost variation ,COVID-19 ,Virology ,Chimeric antigen receptor ,SARS-CoV-2 RNAemia ,AcademicSubjects/MED00290 ,030104 developmental biology ,medicine.anatomical_structure ,Infectious Diseases ,Viral replication ,Viral evolution ,SARS-CoV-2 immune responses ,business ,severe acute respiratory syndrome coronavirus 2 - Abstract
A chimeric antigen receptor-modified T-cell therapy recipient developed severe coronavirus disease 2019, intractable RNAemia, and viral replication lasting >2 months. Premortem endotracheal aspirate contained >2 × 1010 severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA copies/mL and infectious virus. Deep sequencing revealed multiple sequence variants consistent with intrahost virus evolution. SARS-CoV-2 humoral and cell-mediated immunity were minimal. Prolonged transmission from immunosuppressed patients is possible.
- Published
- 2021
24. Interferon-γ: teammate or opponent in the tumour microenvironment?
- Author
-
Angela M. Gocher, Creg J. Workman, and Dario A. A. Vignali
- Subjects
0301 basic medicine ,History ,Cell type ,medicine.medical_treatment ,Article ,Education ,03 medical and health sciences ,Interferon-gamma ,0302 clinical medicine ,Acquired resistance ,Cancer immunotherapy ,Interferon γ ,Neoplasms ,medicine ,Tumor Microenvironment ,Humans ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,Hedgehog signaling pathway ,Computer Science Applications ,Killer Cells, Natural ,030104 developmental biology ,Cytokine ,Cancer research ,business ,030215 immunology - Abstract
Cancer immunotherapy offers substantive benefit to patients with various tumour types, in some cases leading to complete tumour clearance. However, many patients do not respond to immunotherapy, galvanizing the field to define the mechanisms of pre-existing and acquired resistance. Interferon-γ (IFNγ) is a cytokine that has both protumour and antitumour activities, suggesting that it may serve as a nexus for responsiveness to immunotherapy. Many cancer immunotherapies and chemotherapies induce IFNγ production by various cell types, including activated T cells and natural killer cells. Patients resistant to these therapies commonly have molecular aberrations in the IFNγ signalling pathway or express resistance molecules driven by IFNγ. Given that all nucleated cells can respond to IFNγ, the functional consequences of IFNγ production need to be carefully dissected on a cell-by-cell basis. Here, we review the cells that produce IFNγ and the different effects of IFNγ in the tumour microenvironment, highlighting the pleiotropic nature of this multifunctional and abundant cytokine.
- Published
- 2021
25. The costimulatory activity of Tim-3 requires Akt and MAPK signaling and its recruitment to the immune synapse
- Author
-
Judong Lee, Andrea L. Szymczak-Workman, Shunsuke Kataoka, Priyanka Manandhar, Jason Lohmueller, Creg J. Workman, Hridesh Banerjee, Michael Kvorjak, and Lawrence P. Kane
- Subjects
Cell signaling ,Immunological Synapses ,MAP Kinase Signaling System ,T cell ,Lymphocyte Activation ,Biochemistry ,Article ,Immunological synapse ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Hepatitis A Virus Cellular Receptor 2 ,Molecular Biology ,Protein kinase B ,030304 developmental biology ,0303 health sciences ,Chemistry ,T-cell receptor ,Cell Biology ,Chimeric antigen receptor ,Cell biology ,medicine.anatomical_structure ,Phosphorylation ,Persistent Infection ,Signal transduction ,Proto-Oncogene Proteins c-akt ,030215 immunology - Abstract
Expression of the transmembrane protein Tim-3 is increased on dysregulated T cells undergoing chronic activation, including during chronic infection and in solid tumors. Thus, Tim-3 is generally thought of as an inhibitory protein. We and others previously reported that under some circumstances, Tim-3 exerts paradoxical costimulatory activity in T cells (and other cells), including enhancement of the phosphorylation of ribosomal S6 protein. Here, we examined the upstream signaling pathways that control Tim-3-mediated increases in phosphorylated S6 in T cells. We also defined the localization of Tim-3 relative to the T cell immune synapse and its effects on downstream signaling. Recruitment of Tim-3 to the immune synapse was mediated exclusively by the transmembrane domain, replacement of which impaired the ability of Tim-3 to costimulate T cell receptor (TCR)-dependent S6 phosphorylation. Furthermore, enforced localization of the Tim-3 cytoplasmic domain to the immune synapse in a chimeric antigen receptor still enabled T cell activation. Together, our findings are consistent with a model whereby Tim-3 enhances TCR-proximal signaling under acute conditions.
- Published
- 2021
26. Intratumoral regulatory T cells: markers, subsets and their impact on anti‐tumor immunity
- Author
-
Hiroshi Yano, Dario A. A. Vignali, Lawrence P. Andrews, and Creg J. Workman
- Subjects
0301 basic medicine ,Chemokine ,LAG3 ,medicine.medical_treatment ,Immunology ,chemical and pharmacologic phenomena ,T-Lymphocytes, Regulatory ,03 medical and health sciences ,Chemokine receptor ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Review Series: Tregs in Cancer: Where are we now? ,Cancer immunotherapy ,Neoplasms ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Immunology and Allergy ,Tumor microenvironment ,biology ,Immunotherapy ,Phenotype ,030104 developmental biology ,Cytokine ,Cancer research ,biology.protein ,Receptors, Chemokine ,Tumor Escape ,Chemokines ,Inflammation Mediators ,Signal Transduction ,030215 immunology - Abstract
Regulatory T (Treg) cells play a crucial role in maintaining self‐tolerance and resolution of immune responses by employing multifaceted immunoregulatory mechanisms. However, Treg cells readily infiltrate into the tumor microenvironment (TME) and dampen anti‐tumor immune responses, thereby becoming a barrier to effective cancer immunotherapy. There has been a substantial expansion in the development of novel immunotherapies targeting various inhibitory receptors (IRs), such as CTLA4, PD1 and LAG3, but these approaches have mechanistically focused on the elicitation of anti‐tumor responses. However, enhanced inflammation in the TME could also play a detrimental role by facilitating the recruitment, stability and function of Treg cells by up‐regulating chemokines that promote Treg cell migration, and/or increasing inhibitory cytokine production. Furthermore, IR blockade may enhance Treg cell function and survival, thereby serving as a resistance mechanism against effective immunotherapy. Given that Treg cells are comprised of functionally and phenotypically heterogeneous sub‐populations that may alter their characteristics in a context‐dependent manner, it is critical to identify unique molecular pathways that are preferentially used by intratumoral Treg cells. In this review, we discuss markers that serve to identify certain Treg cell subsets, distinguished by chemokine receptors, IRs and cytokines that facilitate their migration, stability and function in the TME. We also discuss how these Treg cell subsets correlate with the clinical outcome of patients with various types of cancer and how they may serve as potential TME‐specific targets for novel cancer immunotherapies.
- Published
- 2019
27. Adaptive plasticity of IL-10+ and IL-35+ Treg cells cooperatively promotes tumor T cell exhaustion
- Author
-
Chang Liu, Robert Lafyatis, Qianxia Zhang, David B. Corry, Mengting Liao, Tracy Tabib, Derrick J Callahan, Tao Sun, Wei Chen, Arjun Pennathur, Dario A. A. Vignali, Hiroshi Yano, Zhe Sun, Maria Chikina, Creg J. Workman, James D. Luketich, Tullia C. Bruno, Amanda C. Poholek, and Deepali V. Sawant
- Subjects
0301 basic medicine ,Adoptive cell transfer ,Tumor microenvironment ,medicine.medical_treatment ,Receptor expression ,T cell ,Immunology ,hemic and immune systems ,chemical and pharmacologic phenomena ,Biology ,Cell biology ,03 medical and health sciences ,Interleukin 10 ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Cancer immunotherapy ,medicine ,Immunology and Allergy ,Memory T cell ,CD8 ,030215 immunology - Abstract
Regulatory T cells (Treg cells) maintain host self-tolerance but are a major barrier to effective cancer immunotherapy. Treg cells subvert beneficial anti-tumor immunity by modulating inhibitory receptor expression on tumor-infiltrating lymphocytes (TILs); however, the underlying mediators and mechanisms have remained elusive. Here, we found that the cytokines IL-10 and IL-35 (Ebi3-IL-12α heterodimer) were divergently expressed by Treg cell subpopulations in the tumor microenvironment (TME) and cooperatively promoted intratumoral T cell exhaustion by modulating several inhibitory receptor expression and exhaustion-associated transcriptomic signature of CD8+ TILs. While expression of BLIMP1 (encoded by Prdm1) was a common target, IL-10 and IL-35 differentially affected effector T cell versus memory T cell fates, respectively, highlighting their differential, partially overlapping but non-redundant regulation of anti-tumor immunity. Our results reveal previously unappreciated cooperative roles for Treg cell-derived IL-10 and IL-35 in promoting BLIMP1-dependent exhaustion of CD8+ TILs that limits effective anti-tumor immunity.
- Published
- 2019
28. Aplp1 and the Aplp1-Lag3 Complex facilitates transmission of pathologic α-synuclein
- Author
-
Xiaobo Mao, Hao Gu, Donghoon Kim, Yasuyoshi Kimura, Ning Wang, Enquan Xu, Haibo Wang, Chan Chen, Shengnan Zhang, Chunyu Jia, Yuqing Liu, Hetao Bian, Senthilkumar S. Karuppagounder, Longgang Jia, Xiyu Ke, Michael Chang, Amanda Li, Jun Yang, Cyrus Rastegar, Manjari Sriparna, Preston Ge, Saurav Brahmachari, Sangjune Kim, Shu Zhang, Yasushi Shimoda, Martina Saar, Creg J. Workman, Dario A. A. Vignali, Ulrike C. Muller, Cong Liu, Han Seok Ko, Valina L. Dawson, and Ted M. Dawson
- Subjects
Alpha-synuclein ,LAG3 ,media_common.quotation_subject ,Neurodegeneration ,Dopaminergic ,Fibril ,medicine.disease ,Cell biology ,chemistry.chemical_compound ,nervous system ,chemistry ,In vivo ,medicine ,APLP1 ,Internalization ,media_common - Abstract
Pathologic α-synuclein (α-syn) spreads from cell-to-cell, in part, through binding to the lymphocyte-activation gene 3 (Lag3). Here we report that amyloid β precursor-like protein 1 (Aplp1) forms a complex with Lag3 that facilitates the binding, internalization, transmission, and toxicity of pathologic α-syn. Deletion of both Aplp1 and Lag3 eliminates the loss of dopaminergic neurons and the accompanying behavioral deficits induced by α-syn preformed fibrils (PFF). Anti-Lag3 prevents the internalization of α-syn PFF by disrupting the interaction of Aplp1 and Lag3, and blocks the neurodegeneration induced by α-syn PFF in vivo. The identification of Aplp1 and the interplay with Lag3 for α-syn PFF induced pathology advances our understanding of the molecular mechanism of cell-to-cell transmission of pathologic α-syn and provides additional targets for therapeutic strategies aimed at preventing neurodegeneration in Parkinson’s disease and related α-synucleinopathies.One Sentence SummaryAplp1 forms a complex with Lag3 that facilitates the binding, internalization, transmission, and toxicity of pathologic α-synuclein.Graphical AbstractAplp1 and the Aplp1-Lag3 complex facilitates transmission of pathologic α-synuclein.Aplp1 is a receptor that drives pathologic α-syn transmission, and genetic depletion of Aplp1 can significantly reduce the α-synuclein pathogenesis. Aplp1 and Lag3 forms an Aplp1-Lag3 complex that accounts for substantial binding of pathologic α-syn to cortical neurons. Together Aplp1 and Lag3 play a major role in pathologic α-syn internalization, transmission and toxicity. Double knockout of Aplp1 and Lag3 and or a Lag3 antibody that disrupts the Aplp1 and Lag3 complex almost completely blocks α-syn PFF-induced neurodegeneration.
- Published
- 2021
29. Bifurcated monocyte states are predictive of mortality in severe COVID-19
- Author
-
Chang Liu, Lawrence C. Andrews, Caleb Lampenfeld, Dario A. A. Vignali, W. Paul Duprex, Alison Morris, Anuradha Ray, Sham Nambulli, Sheryl Kunning, Jishnu Das, Bryan J. McVerry, Georgios D Kitsios, Creg J. Workman, Janet S. Lee, Alok J Joglekar, Ashwin Somasundaram, Carly Cardello, Prabir Ray, Gaurav Deshmukh, Harinder Singh, Barbara Methé, Ayana T. Ruffin, Anthony R. Cillo, Stephanie Grebinowski, Tullia C. Bruno, Panayiotis V. Benos, Sayali Onkar, Yingze Zhang, and Feng Shan
- Subjects
ARDS ,business.industry ,Monocyte ,Disease ,medicine.disease ,Intensive care unit ,Article ,law.invention ,Proinflammatory cytokine ,Immune system ,medicine.anatomical_structure ,Interferon ,law ,Immunology ,Medicine ,CXCL10 ,business ,medicine.drug - Abstract
Coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 infection presents with varied clinical manifestations1, ranging from mild symptoms to acute respiratory distress syndrome (ARDS) with high mortality2,3. Despite extensive analyses, there remains an urgent need to delineate immune cell states that contribute to mortality in severe COVID-19. We performed high-dimensional cellular and molecular profiling of blood and respiratory samples from critically ill COVID-19 patients to define immune cell genomic states that are predictive of outcome in severe COVID-19 disease. Critically ill patients admitted to the intensive care unit (ICU) manifested increased frequencies of inflammatory monocytes and plasmablasts that were also associated with ARDS not due to COVID-19. Single-cell RNAseq (scRNAseq)-based deconvolution of genomic states of peripheral immune cells revealed distinct gene modules that were associated with COVID-19 outcome. Notably, monocytes exhibited bifurcated genomic states, with expression of a cytokine gene module exemplified by CCL4 (MIP-1β) associated with survival and an interferon signaling module associated with death. These gene modules were correlated with higher levels of MIP-1β and CXCL10 levels in plasma, respectively. Monocytes expressing genes reflective of these divergent modules were also detectable in endotracheal aspirates. Machine learning algorithms identified the distinctive monocyte modules as part of a multivariate peripheral immune system state that was predictive of COVID-19 mortality. Follow-up analysis of the monocyte modules on ICU day 5 was consistent with bifurcated states that correlated with distinct inflammatory cytokines. Our data suggests a pivotal role for monocytes and their specific inflammatory genomic states in contributing to mortality in life-threatening COVID-19 disease and may facilitate discovery of new diagnostics and therapeutics.
- Published
- 2021
30. Interferon Gamma Induction of TH1-like Tregs Controls Anti-viral Responses
- Author
-
Angela Marie Gocher, Jian Cui, Andrea Szymczak-Workman, Kate Vignali, Julianna N Latini, Gwen P Pieklo, Lyndsay Avery, Ellyse L Cipolla, Brydie R Huckestein, Lee Hedden, Marlies Meisel, John F Alcorn, Lawrence P Kane, Creg J. Workman, and Dario A. A. Vignali
- Subjects
Immunology ,Immunology and Allergy - Abstract
Regulatory T cells (Tregs) are an immunosuppressive cell population that inhibits immune cell function to maintain peripheral tolerance and homeostasis. We have previously shown that Treg-- response to interferon gamma (IFNγ) is required for response to cancer immunotherapy. We aimed to broaden these findings and determine the impact of Treg response to IFNγ and the IFNγ-inducing cytokine IL12, during viral infection. Mice with Treg-restricted deletion of the IFNγ receptor (IFNGR1, Ifngr1L/LFoxp3Cre-YFP) or the IL12 receptor β2 subunit (IL12Rβ2, Il12b2L/LFoxp3Cre-YFP), were infected with the acute (Armstrong), or chronic (Clone 13), strain of lymphocytic choriomeningitis virus (LCMV). Surprisingly, Treg response to IFNγ but not IL12 induced helper T cell 1 (TH1)-like polarization (expression of T-bet, CXCR3 and IFNγ) of Tregs in mice with chronic LCMV infection. Importantly, this effector-like TH1 Treg state was required for adequate suppression of effector T cells during LCMV infection. Additionally, during LCMV infection, Treg-restricted deletion of IFNGR1 not only prevented TH1-like polarization but also reverted these Tregs to a TH2-like state (expression of GATA-3, CCR4 and IL-4). scRNAseq of viral antigen-specific CD8+ T cells from Clone 13 infected mice showed that Ifngr1-deficient Tregs favored CD8+ T cell memory. These findings were confirmed in an Armstrong/Listeria monocytogenes-GP33 memory model. Further, Ifngr1 deletion from Tregs enhanced response to CD8+ T cell-mediated vaccination when challenged with Clone 13. These findings provide fundamental insight on how Tregs sense inflammatory cues from the environment (IFNγ) during viral infection to prevent overt tissue damage and shape the memory response. Supported by grants from NIH (F32 CA247004-01, T32 CA082084, P01 AI108545, R35 CA263850, R01 CA203689, R01 DK130897, P30 DK120531, P30 CA047904, R01 HL107380, R01 CA206517, R01 AI138504)
- Published
- 2022
31. Regulatory T Cells in the Tumor Microenvironment
- Author
-
Rebekah E, Dadey, Creg J, Workman, and Dario A A, Vignali
- Subjects
Neoplasms ,Interleukin-2 Receptor alpha Subunit ,Tumor Microenvironment ,Humans ,Forkhead Transcription Factors ,T-Lymphocytes, Regulatory - Abstract
Regulatory T cells (T
- Published
- 2020
32. LAG3 associates with TCR-CD3 complexes and suppresses signaling by driving co-receptor-Lck dissociation
- Author
-
Clifford, Guy, Diana M, Mitrea, Po-Chien, Chou, Jamshid, Temirov, Kate M, Vignali, Xueyan, Liu, Hui, Zhang, Richard, Kriwacki, Marcel P, Bruchez, Simon C, Watkins, Creg J, Workman, and Dario A A, Vignali
- Subjects
CD3 Complex ,Antigens, CD ,Lymphocyte Specific Protein Tyrosine Kinase p56(lck) ,CD8 Antigens ,Histocompatibility Antigens Class II ,Receptors, Antigen, T-Cell ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation Gene 3 Protein ,Signal Transduction - Abstract
LAG3 is an inhibitory receptor that is highly expressed on exhausted T cells. Although LAG3-targeting immunotherapeutics are currently in clinical trials, how LAG3 inhibits T cell function remains unclear. Here, we show that LAG3 moved to the immunological synapse and associated with the T cell receptor (TCR)-CD3 complex in CD4
- Published
- 2020
33. A Cre-driven allele-conditioning line to interrogate CD4
- Author
-
Lawrence P, Andrews, Kate M, Vignali, Andrea L, Szymczak-Workman, Amanda R, Burton, Erin A, Brunazzi, Shin Foong, Ngiow, Akihito, Harusato, Arlene H, Sharpe, E John, Wherry, Ichiro, Taniuchi, Creg J, Workman, and Dario A A, Vignali
- Subjects
CD4-Positive T-Lymphocytes ,Gene Editing ,Mice ,Integrases ,Animals ,Cell Differentiation ,Cell Lineage ,CD8-Positive T-Lymphocytes ,Alleles ,Cell Line - Abstract
CD4
- Published
- 2020
34. Resistance to PD1 blockade in the absence of metalloprotease-mediated LAG3 shedding
- Author
-
John M. Kirkwood, Robert L. Ferris, Andrea L. Szymczak-Workman, Ashwin Somasundaram, Chang Liu, Lawrence P. Andrews, Creg J. Workman, Tullia C. Bruno, Evan J. Lipson, Anthony R. Cillo, Jessica M. Moskovitz, Daniel P. Normolle, Kelly D. Moynihan, Darrell J. Irvine, Dario A. A. Vignali, Huang Lin, Ichiro Taniuchi, Massachusetts Institute of Technology. Department of Biological Engineering, and Koch Institute for Integrative Cancer Research
- Subjects
0301 basic medicine ,Male ,LAG3 ,Encephalomyelitis, Autoimmune, Experimental ,Skin Neoplasms ,medicine.medical_treatment ,T cell ,T-Lymphocytes ,Immunology ,Cell ,Programmed Cell Death 1 Receptor ,Melanoma, Experimental ,Mice, Transgenic ,Adenocarcinoma ,Article ,03 medical and health sciences ,ADAM10 Protein ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Antigens, CD ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Immune Checkpoint Inhibitors ,business.industry ,Squamous Cell Carcinoma of Head and Neck ,General Medicine ,Immunotherapy ,Lymphocyte Activation Gene 3 Protein ,Blockade ,030104 developmental biology ,medicine.anatomical_structure ,Drug Resistance, Neoplasm ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Cancer research ,Female ,business ,Transcriptome ,CD8 - Abstract
Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works Mechanisms of resistance to cancer immunotherapy remain poorly understood. Lymphocyte activation gene–3 (LAG3) signaling is regulated by a disintegrin and metalloprotease domain-containing protein–10 (ADAM10)–and ADAM17-mediated cell surface shedding. Here, we show that mice expressing a metalloprotease-resistant, noncleavable LAG3 mutant (LAG3NC) are resistant to PD1 blockade and fail to mount an effective antitumor immune response. Expression of LAG3NC intrinsically perturbs CD4+ T conventional cells (Tconvs), limiting their capacity to provide CD8+ T cell help. Furthermore, the translational relevance for these observations is highlighted with an inverse correlation between high LAG3 and low ADAM10 expression on CD4+ Tconvs in the peripheral blood of patients with head and neck squamous cell carcinoma, which corresponded with poor prognosis. This correlation was also observed in a cohort of patients with skin cancers and was associated with increased disease progression after standard-of-care immunotherapy. These data suggest that subtle changes in LAG3 inhibitory receptor signaling can act as a resistance mechanism with a substantive effect on patient responsiveness to immunotherapy., NIH (Grant EB022433)
- Published
- 2020
35. Regulatory T Cells in the Tumor Microenvironment
- Author
-
Creg J. Workman, Rebekah E. Dadey, and Dario A. A. Vignali
- Subjects
Tumor microenvironment ,LAG3 ,medicine.medical_treatment ,FOXP3 ,hemic and immune systems ,chemical and pharmacologic phenomena ,Biology ,03 medical and health sciences ,Interleukin 10 ,0302 clinical medicine ,Cancer immunotherapy ,Cancer research ,medicine ,030212 general & internal medicine ,IL-2 receptor ,Receptor ,Transcription factor - Abstract
Regulatory T cells (Tregs) are an immunosuppressive subpopulation of CD4+ T cells that are endowed with potent suppressive activity and function to limit immune activation and maintain homeostasis. These cells are identified by the hallmark transcription factor FOXP3 and the high-affinity interleukin-2 (IL-2) receptor chain CD25. Tregs can be recruited to and persist within the tumor microenvironment (TME), acting as a potent barrier to effective antitumor immunity. This chapter will discuss [i] the history and hallmarks of Tregs; [ii] the recruitment, development, and persistence of Tregs within the TME; [iii] Treg function within TME; asnd [iv] the therapeutic targeting of Tregs in the clinic. This chapter will conclude with a discussion of likely trends and future directions.
- Published
- 2020
36. The co-stimulatory activity of Tim-3 requires Akt and MAPK signaling and immune synapse recruitment
- Author
-
Shunsuke Kataoka, Priyanka Manandhar, Judong Lee, Creg J. Workman, Hridesh Banerjee, Andrea L. Szymczak-Workman, Michael Kvorjak, Jason Lohmueller, and Lawrence P. Kane
- Subjects
0303 health sciences ,Chemistry ,T cell ,Chimeric antigen receptor ,Transmembrane protein ,3. Good health ,Immunological synapse ,Cell biology ,03 medical and health sciences ,Transmembrane domain ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Protein phosphorylation ,Signal transduction ,Protein kinase B ,030304 developmental biology ,030215 immunology - Abstract
Expression of the transmembrane protein Tim-3 is increased on dysregulated T cells undergoing chronic T cell activation, including in chronic infection and solid tumors. We and others previously reported that Tim-3 exerts apparently paradoxical co-stimulatory activity in T cells (and other cells), including enhancement of ribosomal S6 protein phosphorylation (pS6). Here we examined the upstream signaling pathways that control Tim3-mediated increases in pS6 in T cells. We have also defined the localization of Tim-3 relative to the T cell immune synapse and impacts on downstream signaling. Recruitment of Tim-3 to the immune synapse was mediated exclusively by the transmembrane domain, replacement of which impaired Tim-3 co-stimulation of pS6. Strikingly, enforced localization of the Tim-3 cytoplasmic domain to the immune synapse in the context of a chimeric antigen receptor still allowed for robust T cell activation. Our findings are consistent with a model whereby Tim-3 enhances TCR-proximal signaling under acute conditions.One Sentence SummaryHere we define elements of signaling and localization associated with Tim-3 co-stimulatory function in T cells.
- Published
- 2019
37. People critically ill with COVID-19 exhibit peripheral immune profiles predictive of mortality and reflective of SARS-CoV-2 lung viral burden
- Author
-
Anthony R. Cillo, Ashwin Somasundaram, Feng Shan, Carly Cardello, Creg J. Workman, Georgios D. Kitsios, Ayana T. Ruffin, Sheryl Kunning, Caleb Lampenfeld, Sayali Onkar, Stephanie Grebinoski, Gaurav Deshmukh, Barbara Methe, Chang Liu, Sham Nambulli, Lawrence P. Andrews, W. Paul Duprex, Alok V. Joglekar, Panayiotis V. Benos, Prabir Ray, Anuradha Ray, Bryan J. McVerry, Yingze Zhang, Janet S. Lee, Jishnu Das, Harinder Singh, Alison Morris, Tullia C. Bruno, and Dario A.A. Vignali
- Subjects
Inflammation ,SARS-CoV-2 ,inflammatory cytokines ,Critical Illness ,gene modules ,inflammatory monocytes ,COVID-19 ,Reproducibility of Results ,severe COVID-19 ,Models, Theoretical ,Viral Load ,Monocytes ,General Biochemistry, Genetics and Molecular Biology ,machine learning ,Report ,single-cell RNAseq ,Cytokines ,Humans ,type I interferon ,Gene Regulatory Networks ,Myeloid Cells ,COVID-19 outcome ,Lung ,Aged - Abstract
Despite extensive analyses, there remains an urgent need to delineate immune cell states that contribute to mortality in critically ill Coronavirus disease 2019 (COVID-19) patients. Here, we present high-dimensional profiling of blood and respiratory samples in severe COVID-19 patients to examine the association between cell-linked molecular features and mortality outcomes. Peripheral transcriptional profiles by single-cell RNAseq based deconvolution of immune states are associated with COVID-19 mortality. Further, persistently high levels of an interferon signaling module in monocytes over time leads to subsequent concerted upregulation of inflammatory cytokines. SARS-CoV-2 infected myeloid cells in the lower respiratory tract upregulate CXCL10, leading to a higher risk of death. Our analysis suggests a pivotal role for viral infected myeloid cells and protracted interferon signaling in severe COVID-19., Graphical Abstract, Cillo et al identify transcriptional profiles in peripheral blood that are associated with mortality in critically ill COVID-19 patients. Inflammatory monocyte signatures are correlated with CXCL10 in plasma and precede upregulation of inflammatory cytokines in blood. SARS-CoV-2-infected macrophages in the respiratory tract expressed CXCL10, linking peripheral and lung immune profiles.
- Published
- 2021
38. 253 PD1 and LAG3 converge to limit polyfunctionality and systemic immunity
- Author
-
Dario A. A. Vignali, E. John Wherry, Creg J. Workman, Lawrence C. Andrews, and Sasikanth Manne
- Subjects
Pharmacology ,Physics ,Cancer Research ,LAG3 ,Oncology ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Molecular Medicine ,Immunology and Allergy ,Systemic immunity ,Limit (mathematics) ,RC254-282 - Abstract
BackgroundTargeting PD1 with monoclonal antibodies has yielded clinical success across a variety of tumor types, yet overcoming inhibitory receptor (IR)-mediated tolerance is essential to improve immunotherapeutic responses. LAG3 co-expresses with PD1 on CD8+ tumor-infiltrating T cells (TIL), signifying a highly exhausted phenotype, and dual PD1/LAG3 blockade in C57BL/6 mice enhances antitumor immunity. As CD8+ TIL is the dominant LAG3-expressing TIL population, it is hypothesized that PD1 and LAG3 synergizes to limit CD8+ TIL function controlling antitumor immunity.MethodsTo understand the cellular and mechanistic basis for PD1/LAG3 synergy, conditional knock-in mice ”surgically dissect” Pdcd1 and/or Lag3 floxed alleles restricted to CD8+ T cells expressing E8ICre.GFP. These mice were crossed with the Pmel transgene to assess PD1 and/or LAG3-sufficient or deficient gp100-specific CD8+ T cell populations. CD8+ Pmel cells were isolated and adoptively transferred into C57BL/6 mice harboring a B16-gp100-overexpressing tumor to observe therapeutic benefit, or to assess T cell functionality within the tumor.ResultsWhile little therapeutic benefit was observed with a prophylactic adoptive transfer of wild-type CD8+ Pmel cells into mice which then received B16-gp100 tumor, there was reduced tumor growth in mice receiving PD1-deficient CD8+ Pmel cells, which was further enhanced in mice receiving PD1/LAG3-deficient CD8+ Pmel cells with long-term tumor-free survival. Likewise, a therapeutic administration of PD1/LAG3-deficient CD8+ Pmel cells into mice once the tumor has been established showed an initial therapeutic benefit, with enhanced survival, that was not demonstrated with adoptive transfer of PD1 or LAG3-deficient, or wild-type, counterparts. Each PD1 and/or LAG3-sufficient or deficient CD8+ Pmel mice were differentially congenically marked to assess each of the four genotypes that can be adoptively transferred into the same host as a ”quad transfer” system. Recovery of these populations within the tumor show that the PD1/LAG3-deficient CD8+ Pmel cells out-compete PD1 or LAG3-deficient, or wild-type, counterparts due to enhanced proliferation (Ki67/BrdU). Furthermore, PD1/LAG3-deficient CD8+ T cells were more functional with increased IFNg and GzmB release observed by flow cytometry.ConclusionsOverall PD1 and LAG3 limit anti-tumor immune effects as removal of both IRs on a gp100 antigen-specific CD8+ T cell population results in reduced B16-gp100 tumor growth and enhanced survival in an adoptive transfer model, as a result of enhanced CD8+ TIL functionality and proliferation. These results provide striking evidence that the development of anti-LAG3 agents in the clinic would yield improved responses with anti-PD1.
- Published
- 2021
39. Neuropilin-1 is a T cell memory checkpoint limiting long-term antitumor immunity
- Author
-
Chang, Liu, Ashwin, Somasundaram, Sasikanth, Manne, Angela M, Gocher, Andrea L, Szymczak-Workman, Kate M, Vignali, Ellen N, Scott, Daniel P, Normolle, E, John Wherry, Evan J, Lipson, Robert L, Ferris, Tullia C, Bruno, Creg J, Workman, and Dario A A, Vignali
- Subjects
Mice, Knockout ,Precursor Cells, T-Lymphoid ,Programmed Cell Death 1 Receptor ,Immunity ,Melanoma, Experimental ,CD8-Positive T-Lymphocytes ,Immune Checkpoint Proteins ,Neuropilin-1 ,Mice ,Cell Line, Tumor ,Immune Tolerance ,Animals ,Humans ,Immunologic Memory ,Signal Transduction - Abstract
Robust CD8
- Published
- 2019
40. Lymphocyte-Activation Gene 3 (LAG3): the Next Immune Checkpoint Receptor
- Author
-
Elisa Ruffo, Richard Wu, Tullia C. Bruno, Creg J. Workman, and Dario A. A. Vignali
- Subjects
0301 basic medicine ,LAG3 ,T cell ,medicine.medical_treatment ,Immunology ,Ligands ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Antigens, CD ,Neoplasms ,Tumor Microenvironment ,Immunology and Allergy ,Medicine ,Animals ,Humans ,Receptors, Immunologic ,business.industry ,Cancer ,medicine.disease ,Lymphocyte Activation Gene 3 Protein ,Immune checkpoint ,Chronic infection ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Immunotherapy ,business ,CD8 - Abstract
Immune checkpoint therapy has revolutionized cancer treatment by blocking inhibitory pathways in T cells that limits the an effective anti-tumor immune response. Therapeutics targeting CTLA-4 and PD1/PDL1 have progressed to first line therapy in multiple tumor types with some patients exhibiting tumor regression or remission. However, the majority of patients do not benefit from checkpoint therapy emphasizing the need for alternative therapeutic options. Lymphocyte Activation Gene 3 (LAG3) or CD223 is expressed on multiple cell types including CD4(+) and CD8(+) T cells, and T(regs), and is required for optimal T cell regulation and homeostasis. Persistent antigen-stimulation in cancer or chronic infection leads to chronic LAG3 expression, promoting T cell exhaustion. Targeting LAG3 along with PD1 facilitates T cell reinvigoration. A substantial amount of pre-clinical data and mechanistic analysis has led to LAG3 being the third checkpoint to be targeted in the clinic with nearly a dozen therapeutics under investigation. In this review, we will discuss the structure, function and role of LAG3 in murine and human models of disease, including autoimmune and inflammatory diseases, chronic viral and parasitic infections, and cancer, emphasizing new advances in the development of LAG3-targeting immunotherapies for cancer that are currently in clinical trials.
- Published
- 2019
41. Adaptive plasticity of IL-10
- Author
-
Deepali V, Sawant, Hiroshi, Yano, Maria, Chikina, Qianxia, Zhang, Mengting, Liao, Chang, Liu, Derrick J, Callahan, Zhe, Sun, Tao, Sun, Tracy, Tabib, Arjun, Pennathur, David B, Corry, James D, Luketich, Robert, Lafyatis, Wei, Chen, Amanda C, Poholek, Tullia C, Bruno, Creg J, Workman, and Dario A A, Vignali
- Subjects
Immunity, Cellular ,Gene Expression Profiling ,Interleukins ,Melanoma, Experimental ,Adoptive Transfer ,T-Lymphocytes, Regulatory ,Interleukin-10 ,Mice ,Neoplasms ,Animals ,Cytokines ,Humans ,Transcriptome ,Signal Transduction - Abstract
Regulatory T cells (T
- Published
- 2018
42. Competition for Active TGFβ Cytokine Allows for Selective Retention of Antigen-Specific Tissue- Resident Memory T Cells in the Epidermal Niche
- Author
-
Dario A. A. Vignali, David W. Griggs, Daniel H. Kaplan, Haiyue Li, Creg J. Workman, Yi Yang, Toshiro Hirai, Yukari Zenke, David Masopust, Breanna Anh Thu Nguyen, Harinder Singh, Laurent Bartholin, Jacinto S. De La Cruz Diaz, Virendra K. Chaudhri, and Paul Yifan Zhou
- Subjects
Keratinocytes ,0301 basic medicine ,Receptors, Antigen, T-Cell, alpha-beta ,medicine.medical_treatment ,Immunology ,T-Cell Antigen Receptor Specificity ,CD8-Positive T-Lymphocytes ,Biology ,Binding, Competitive ,T-Lymphocytes, Regulatory ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Transforming Growth Factor beta ,medicine ,Bystander effect ,Animals ,Immunology and Allergy ,Autocrine signalling ,Lymph node ,integumentary system ,Epidermis (botany) ,Bystander Effect ,Clone Cells ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Infectious Diseases ,Cytokine ,medicine.anatomical_structure ,Cellular Microenvironment ,Organ Specificity ,030220 oncology & carcinogenesis ,Epidermis ,Immunologic Memory ,CD8 ,Signal Transduction ,Transforming growth factor - Abstract
Summary Following antigen-driven expansion in lymph node, transforming growth factor-β (TGFβ) is required for differentiation of skin-recruited CD8+ T cell effectors into epidermal resident memory T (Trm) cells and their epidermal persistence. We found that the source of TGFβ -supporting Trm cells was autocrine. In addition, antigen-specific Trm cells that encountered cognate antigen in the skin, and bystander Trm cells that did not, both displayed long-term persistence in the epidermis under steady-state conditions. However, when the active-TGFβ was limited or when new T cell clones were recruited into the epidermis, antigen-specific Trm cells were more efficiently retained than bystander Trm cells. Genetically enforced TGFβR signaling allowed bystander Trm cells to persist in the epidermis as efficiently as antigen-specific Trm cells in both contexts. Thus, competition between T cells for active TGFβ represents an unappreciated selective pressure that promotes the accumulation and persistence of antigen-specific Trm cells in the epidermal niche.
- Published
- 2021
43. Interleukin-35 Limits Anti-Tumor Immunity
- Author
-
Meghan E. Turnis, Hiroshi Yano, Lawrence P. Andrews, Amy J. Beres, Deepali V. Sawant, Creg J. Workman, Dario A. A. Vignali, Greg M. Delgoffe, Andrea L. Szymczak-Workman, and Peter Vogel
- Subjects
0301 basic medicine ,Skin Neoplasms ,LAG3 ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Immunology ,Melanoma, Experimental ,chemical and pharmacologic phenomena ,Cell Growth Processes ,T-Lymphocytes, Regulatory ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigens, CD ,Immunity ,Cell Line, Tumor ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Immunology and Allergy ,Antibodies, Blocking ,Hepatitis A Virus Cellular Receptor 2 ,Cell Proliferation ,Mice, Knockout ,Tumor microenvironment ,biology ,Antitumor immunity ,Effector ,Interleukins ,Lymphocyte Activation Gene 3 Protein ,Tumor Burden ,Mice, Inbred C57BL ,030104 developmental biology ,Infectious Diseases ,Cytokine ,030220 oncology & carcinogenesis ,Interleukin 35 ,biology.protein ,Receptors, Virus ,Antibody ,Immunologic Memory - Abstract
Summary Regulatory T (Treg) cells pose a major barrier to effective anti-tumor immunity. Although Treg cell depletion enhances tumor rejection, the ensuing autoimmune sequelae limits its utility in the clinic and highlights the need for limiting Treg cell activity within the tumor microenvironment. Interleukin-35 (IL-35) is a Treg cell-secreted cytokine that inhibits T cell proliferation and function. Using an IL-35 reporter mouse, we observed substantial enrichment of IL-35 + Treg cells in tumors. Neutralization with an IL-35-specific antibody or Treg cell-restricted deletion of IL-35 production limited tumor growth in multiple murine models of human cancer. Limiting intratumoral IL-35 enhanced T cell proliferation, effector function, antigen-specific responses, and long-term T cell memory. Treg cell-derived IL-35 promoted the expression of multiple inhibitory receptors (PD1, TIM3, LAG3), thereby facilitating intratumoral T cell exhaustion. These findings reveal previously unappreciated roles for IL-35 in limiting anti-tumor immunity and contributing to T cell dysfunction in the tumor microenvironment.
- Published
- 2016
44. Neuropilin-1: a checkpoint target with unique implications for cancer immunology and immunotherapy
- Author
-
Tullia C. Bruno, Chang Liu, Dario A. A. Vignali, Creg J. Workman, and Christopher A. Chuckran
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,T cell ,Immunology ,Review ,immunomodulation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Neoplasms ,Animals ,Humans ,tumor microenvironment ,Immunology and Allergy ,Medicine ,RC254-282 ,Cancer immunology ,Pharmacology ,Tumor microenvironment ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Peripheral tolerance ,Immunotherapy ,Neuropilin-1 ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Cancer research ,Molecular Medicine ,Female ,immunotherapy ,business ,Checkpoint Blockade Immunotherapy ,CD8 ,030215 immunology - Abstract
Checkpoint blockade immunotherapy established a new paradigm in cancer treatment: for certain patients curative treatment requires immune reinvigoration. Despite this monumental advance, only 20%–30% of patients achieve an objective response to standard of care immunotherapy, necessitating the consideration of alternative targets. Optimal strategies will not only stimulate CD8+T cells, but concomitantly modulate immunosuppressive cells in the tumor microenvironment (TME), most notably regulatory T cells (Tregcells). In this context, the immunoregulatory receptor Neuropilin-1 (NRP1) is garnering renewed attention as it reinforces intratumoral Tregcell function amidst inflammation in the TME. Loss of NRP1 on Tregcells in mouse models restores antitumor immunity without sacrificing peripheral tolerance. Enrichment of NRP1+Tregcells is observed in patients across multiple malignancies with cancer, both intratumorally and in peripheral sites. Thus, targeting NRP1 may safely undermine intratumoral Tregcell fitness, permitting enhanced inflammatory responses with existing immunotherapies. Furthermore, NRP1 has been recently found to modulate tumor-specific CD8+T cell responses. Emerging data suggest that NRP1 restricts CD8+T cell reinvigoration in response to checkpoint inhibitors, and more importantly, acts as a barrier to the long-term durability of CD8+T cell-mediated tumor immunosurveillance. These novel and distinct regulatory mechanisms present an exciting therapeutic opportunity. This review will discuss the growing literature on NRP1-mediated immune modulation which provides a strong rationale for categorizing NRP1 as both a key checkpoint in the TME as well as an immunotherapeutic target with promise either alone or in combination with current standard of care therapeutic regimens.
- Published
- 2020
45. Interferon gamma production by regulatory T cells is required for response to cancer immunotherapy
- Author
-
Angela Marie Gocher, Sanah Handu, Creg J Workman, and Dario A. A. Vignali
- Subjects
Immunology ,Immunology and Allergy - Abstract
We have shown that murine regulatory T cells (Tregs) produce the cytokine interferon gamma (IFNγ) during anti-PD1 immunotherapy and that IFNγ-driven Treg dysfunction is necessary for tumor clearance by anti-PD1. However, it is unknown how Treg production of IFNγ dictates response to cancer immunotherapy. It has been shown that IL12-induced production of IFNγ is required for response to anti-PD1 therapy. However, it has yet to be determined whether Tregs are a key responder to IL12. Thus, elucidating the interplay of IL12, IFNγ and Tregs in the tumor microenvironment (TME) is essential for understanding the mechanistic events required for response to immunotherapy. Our lab has generated two novel murine models that allow for Treg-restricted deletion of Ifng (IfngL/LFoxp3Cre-YFP) or Il12rb2 (Il12rb2L/LFoxp3Cre-YFP). These murine models were used to assess the contribution of Treg-derived IFNγ and response to IL12 on the growth of various syngeneic tumor models and response to checkpoint blockade, vaccination and tumor-specific antibodies. Surprisingly, mice with Ifng-deficient Tregs were completely resistant to anti-PD1 immunotherapy. Preliminary experiments suggest that IFNγ production by Tregs is an early requirement to produce an effector-like Treg state in order to mediate an effective anti-tumor response. Mice with Il12rb2-deficient Tregs had diminished therapeutic response to anti-PD1. These data suggest that IFNγ producing Tregs are required to shift the balance from an immunosuppressive TME to one that favors the reinvigoration of the anti-tumor response generated by cancer immunotherapies. Future studies will determine the mechanisms of resistance and whether IFNγ+ Tregs predict response to immunotherapy.
- Published
- 2020
46. Treg Cells Promote the SREBP1-Dependent Metabolic Fitness of Tumor-Promoting Macrophages via Repression of CD8
- Author
-
Chang, Liu, Maria, Chikina, Rahul, Deshpande, Ashley V, Menk, Ting, Wang, Tracy, Tabib, Erin A, Brunazzi, Kate M, Vignali, Ming, Sun, Donna B, Stolz, Robert A, Lafyatis, Wei, Chen, Greg M, Delgoffe, Creg J, Workman, Stacy G, Wendell, and Dario A A, Vignali
- Subjects
Mice, Knockout ,Carcinogenesis ,Macrophages ,Fatty Acids ,Melanoma, Experimental ,chemical and pharmacologic phenomena ,Cell Differentiation ,Forkhead Transcription Factors ,Neoplasms, Experimental ,CD8-Positive T-Lymphocytes ,T-Lymphocytes, Regulatory ,Neuropilin-1 ,Article ,Mice, Inbred C57BL ,Interferon-gamma ,Mice ,Th2 Cells ,Tumor Microenvironment ,Animals ,Sterol Regulatory Element Binding Protein 1 ,Melanoma ,Immune Evasion - Abstract
Regulatory T (Treg) cells are crucial for immune homeostasis but they also contribute to tumor immune evasion by promoting a suppressive tumor microenvironment (TME). Mice with Treg cell-restricted Neuropilin 1 deficiency show tumor resistance while maintaining peripheral immune homeostasis, thereby providing a controlled system to interrogate the impact of intratumoral Treg cells on the TME. Using this and other genetic models, we showed that Treg cells shaped the transcriptional landscape across multiple tumor-infiltrating immune cell types. Treg cells suppressed CD8(+) T cell secretion of interferon-γ (IFNγ), which would otherwise block the activation of sterol regulatory elementbinding protein 1 (SREBP1)-mediated fatty acid synthesis in immunosuppressive (M2-like) tumor-associated macrophages (TAMs). Thus, Treg cells indirectly but selectively sustained M2-like TAM metabolic fitness, mitochondrial integrity and survival. SREBP1 inhibition augmented the efficacy of immune checkpoint blockade, suggesting that targeting Treg cells or their modulation of lipid metabolism in M2-like TAMs could improve cancer immunotherapy.
- Published
- 2018
47. Identification of the Docking Site for CD3 on the T Cell Receptor β Chain by Solution NMR
- Author
-
Roy A. Mariuzza, Ming Luo, Yihong Chen, Yiyuan Yin, John Orban, Qian Wang, Kate M. Vignali, Dario A. A. Vignali, Melissa C. Kerzic, Creg J. Workman, Sneha Rangarajan, and Yanan He
- Subjects
Protein Folding ,Magnetic Resonance Spectroscopy ,CD3 Complex ,Receptors, Antigen, T-Cell, alpha-beta ,T-Lymphocytes ,Protein subunit ,T cell ,CD3 ,Molecular Sequence Data ,Immunology ,Gene Expression ,chemical and pharmacologic phenomena ,Biology ,Biochemistry ,Molecular Docking Simulation ,Protein Structure, Secondary ,Escherichia coli ,medicine ,Humans ,Amino Acid Sequence ,Molecular Biology ,T-cell receptor ,hemic and immune systems ,Cell Biology ,Recombinant Proteins ,Protein Structure, Tertiary ,Protein Subunits ,medicine.anatomical_structure ,Structural biology ,Receptor-CD3 Complex, Antigen, T-Cell ,Docking (molecular) ,Mutation ,Biophysics ,biology.protein ,Protein Multimerization ,Signal transduction - Abstract
The T cell receptor (TCR)-CD3 complex is composed of a genetically diverse αβ TCR heterodimer associated noncovalently with the invariant CD3 dimers CD3ϵγ, CD3ϵδ, and CD3ζζ. The TCR mediates peptide-MHC recognition, whereas the CD3 molecules transduce activation signals to the T cell. Although much is known about downstream T cell signaling pathways, the mechanism whereby TCR engagement by peptide-MHC initiates signaling is poorly understood. A key to solving this problem is defining the spatial organization of the TCR-CD3 complex and the interactions between its subunits. We have applied solution NMR methods to identify the docking site for CD3 on the β chain of a human autoimmune TCR. We demonstrate a low affinity but highly specific interaction between the extracellular domains of CD3 and the TCR constant β (Cβ) domain that requires both CD3ϵγ and CD3ϵδ subunits. The mainly hydrophilic docking site, comprising 9-11 solvent-accessible Cβ residues, is relatively small (∼400 Å(2)), consistent with the weak interaction between TCR and CD3 extracellular domains, and devoid of glycosylation sites. The docking site is centered on the αA and αB helices of Cβ, which are located at the base of the TCR. This positions CD3ϵγ and CD3ϵδ between the TCR and the T cell membrane, permitting us to distinguish among several possible models of TCR-CD3 association. We further correlate structural results from NMR with mutational data on TCR-CD3 interactions from cell-based assays.
- Published
- 2015
48. LAG-3 limits regulatory T cell proliferation and function in autoimmune diabetes
- Author
-
Dario A. A. Vignali, Andrea L. Szymczak-Workman, Kate M. Vignali, Maria Chikina, William Horne, Jay K. Kolls, Daniel P. Normolle, Maria Bettini, Creg J. Workman, and Qianxia Zhang
- Subjects
0301 basic medicine ,Autoimmune disease ,LAG3 ,Regulatory T cell ,Activator (genetics) ,Cell growth ,Effector ,Immunology ,Cell ,hemic and immune systems ,chemical and pharmacologic phenomena ,General Medicine ,Biology ,medicine.disease ,medicine.disease_cause ,Article ,Autoimmunity ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,medicine - Abstract
Inhibitory receptors are pivotal in controlling T cell homeostasis because of their intrinsic regulation of conventional effector T (Tconv) cell proliferation, viability and function. However, the role of Inhibitory receptors on regulatory T (Treg) cells remains obscure, as they could be required for suppressive activity and/or limit Treg cell function. We evaluated the role of Lymphocyte Activation Gene-3 (LAG3, CD223) on Treg cells by generating mice in which LAG3 is absent on the cell surface of Treg cells in a murine model of Type 1 Diabetes. Surprisingly, mice that lacked LAG3 expression on Treg cells exhibited reduced autoimmune diabetes, consistent with enhanced Treg cell proliferation and function. Whereas the transcriptional landscape of peripheral wild-type (WT) and Lag3-deficient Treg cells was largely comparable, substantial differences between intra-islet Treg cells were evident and involved a subset of genes and pathways that promote Treg cell maintenance and function. Consistent with these observations, Lag3-deficient Treg cells out-competed WT Treg cells in the islets but not in the periphery in co-transfer experiments due to enhanced IL2-Stat5 signaling and increased Eos expression. Our study suggests that LAG3 intrinsically limits Treg cell proliferation and function at inflammatory sites, promotes autoimmunity in a chronic autoimmune-prone environment and may contribute to Treg cell insufficiency in autoimmune disease.
- Published
- 2017
49. Treg Cells Promote the SREBP1-Dependent Metabolic Fitness of Tumor-Promoting Macrophages via Repression of CD8+ T Cell-Derived Interferon-γ
- Author
-
Greg M. Delgoffe, Maria Chikina, Chang Liu, Tracy Tabib, Ashley V. Menk, Creg J. Workman, Stacy G. Wendell, Rahul Deshpande, Donna B. Stolz, Ming Sun, Erin A. Brunazzi, Robert Lafyatis, Dario A. A. Vignali, Kate M. Vignali, Ting Wang, and Wei Chen
- Subjects
0301 basic medicine ,Cell type ,Tumor microenvironment ,medicine.medical_treatment ,Immunology ,chemical and pharmacologic phenomena ,Biology ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,Immune system ,Cancer immunotherapy ,030220 oncology & carcinogenesis ,Genetic model ,medicine ,Cancer research ,Immunology and Allergy ,Cytotoxic T cell ,CD8 - Abstract
Summary Regulatory T (Treg) cells are crucial for immune homeostasis, but they also contribute to tumor immune evasion by promoting a suppressive tumor microenvironment (TME). Mice with Treg cell-restricted Neuropilin-1 deficiency show tumor resistance while maintaining peripheral immune homeostasis, thereby providing a controlled system to interrogate the impact of intratumoral Treg cells on the TME. Using this and other genetic models, we showed that Treg cells shaped the transcriptional landscape across multiple tumor-infiltrating immune cell types. Treg cells suppressed CD8+ T cell secretion of interferon-γ (IFNγ), which would otherwise block the activation of sterol regulatory element-binding protein 1 (SREBP1)-mediated fatty acid synthesis in immunosuppressive (M2-like) tumor-associated macrophages (TAMs). Thus, Treg cells indirectly but selectively sustained M2-like TAM metabolic fitness, mitochondrial integrity, and survival. SREBP1 inhibition augmented the efficacy of immune checkpoint blockade, suggesting that targeting Treg cells or their modulation of lipid metabolism in M2-like TAMs could improve cancer immunotherapy.
- Published
- 2019
50. Interferon-γ Drives T
- Author
-
Abigail E, Overacre-Delgoffe, Maria, Chikina, Rebekah E, Dadey, Hiroshi, Yano, Erin A, Brunazzi, Gulidanna, Shayan, William, Horne, Jessica M, Moskovitz, Jay K, Kolls, Cindy, Sander, Yongli, Shuai, Daniel P, Normolle, John M, Kirkwood, Robert L, Ferris, Greg M, Delgoffe, Tullia C, Bruno, Creg J, Workman, and Dario A A, Vignali
- Subjects
Male ,Gene Expression Profiling ,Programmed Cell Death 1 Receptor ,Melanoma, Experimental ,Forkhead Transcription Factors ,T-Lymphocytes, Regulatory ,Neuropilin-1 ,Mice, Inbred C57BL ,Interferon-gamma ,Mice ,Head and Neck Neoplasms ,Carcinoma, Squamous Cell ,Tumor Microenvironment ,Animals ,Humans ,Female ,Gene Regulatory Networks ,Melanoma ,Receptors, Interferon - Abstract
Regulatory T cells (T
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.