35 results on '"Dogra, Pranay"'
Search Results
2. Site-specific development and progressive maturation of human tissue-resident memory T cells over infancy and childhood
- Author
-
Connors, Thomas J., Matsumoto, Rei, Verma, Shivali, Szabo, Peter A., Guyer, Rebecca, Gray, Joshua, Wang, Zicheng, Thapa, Puspa, Dogra, Pranay, Poon, Maya M.L., Rybkina, Ksenia, Bradley, Marissa C., Idzikowski, Emma, McNichols, James, Kubota, Masaru, Pethe, Kalpana, Shen, Yufeng, Atkinson, Mark A., Brusko, Maigan, Brusko, Todd M., Yates, Andrew J., Sims, Peter A., and Farber, Donna L.
- Published
- 2023
- Full Text
- View/download PDF
3. Inhaled particulate accumulation with age impairs immune function and architecture in human lung lymph nodes
- Author
-
Ural, Basak B., Caron, Daniel P., Dogra, Pranay, Wells, Steven B., Szabo, Peter A., Granot, Tomer, Senda, Takashi, Poon, Maya M. L., Lam, Nora, Thapa, Puspa, Lee, Yoon Seung, Kubota, Masaru, Matsumoto, Rei, and Farber, Donna L.
- Published
- 2022
- Full Text
- View/download PDF
4. Distinct antibody responses to SARS-CoV-2 in children and adults across the COVID-19 clinical spectrum
- Author
-
Weisberg, Stuart P., Connors, Thomas J., Zhu, Yun, Baldwin, Matthew R., Lin, Wen-Hsuan, Wontakal, Sandeep, Szabo, Peter A., Wells, Steven B., Dogra, Pranay, Gray, Joshua, Idzikowski, Emma, Stelitano, Debora, Bovier, Francesca T., Davis-Porada, Julia, Matsumoto, Rei, Poon, Maya Meimei Li, Chait, Michael, Mathieu, Cyrille, Horvat, Branka, Decimo, Didier, Hudson, Krystalyn E., Zotti, Flavia Dei, Bitan, Zachary C., La Carpia, Francesca, Ferrara, Stephen A., Mace, Emily, Milner, Joshua, Moscona, Anne, Hod, Eldad, Porotto, Matteo, and Farber, Donna L.
- Published
- 2021
- Full Text
- View/download PDF
5. Intrathymic differentiation of natural antibody-producing plasma cells in human neonates
- Author
-
Cordero, Hector, King, Rodney G., Dogra, Pranay, Dufeu, Chloe, See, Sarah B., Chong, Alexander M., Uhlemann, Anne-Catrin, Ho, Siu-Hong, Kalfa, David M., Bacha, Emile A., Kearney, John F., and Zorn, Emmanuel
- Published
- 2021
- Full Text
- View/download PDF
6. Efbalropendekin Alfa enhances human natural killer cell cytotoxicity against tumor cell lines in vitro.
- Author
-
Shehata, Hesham M., Dogra, Pranay, Gierke, Sarah, Holder, Patrick, and Sanjabi, Shomyseh
- Subjects
KILLER cells ,ANTIBODY-dependent cell cytotoxicity ,CYTOTOXINS ,CELL lines ,KRA - Abstract
IL-15 has shown preclinical activity by enhancing the functional maturation of natural killer (NK) cells. Clinical evaluation of the potential anticancer activity of most cytokines, including IL-15, has been limited by low tolerability and rapid in vivo clearance. Efbalropendekin Alfa (XmAb24306) is a soluble IL15/IL15-receptor alpha heterodimer complex fused to a half-life extended Fc domain (IL15/IL15Ra-Fc), engineered with mutations to reduce IL-15 affinity for CD122. Reduced affinity drives lower potency, leading to prolonged pharmacodynamic response in cynomolgus monkeys. We show that in vitro, human NK cells treated with XmAb24306 demonstrate enhanced cytotoxicity against various tumor cell lines. XmAb24306-treated NK cells also exhibit enhanced killing of 3D colorectal cancer spheroids. Daratumumab (dara), a monoclonal antibody (mAb) that targets CD38 results in antibody-dependent cellular cytotoxicity (ADCC) of both multiple myeloma (MM) cells and NK cells. Addition of XmAb24306 increases dara-mediated NK cell ADCC against various MM cell lines in vitro. Because NK cells express CD38, XmAb24306 increases dara-mediated NK cell fratricide, but overall does not negatively impact the ADCC activity against a MM cell line likely due to increased NK cell activity of the surviving cells. These data show that XmAb24306 increases direct and ADCC-mediated human NK cell cytotoxicity in vitro. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Microanatomical dissection of human intestinal T-cell immunity reveals site-specific changes in gut-associated lymphoid tissues over life
- Author
-
Senda, Takashi, Dogra, Pranay, Granot, Tomer, Furuhashi, Kazuhiro, Snyder, Mark E., Carpenter, Dustin J., Szabo, Peter A., Thapa, Puspa, Miron, Michelle, and Farber, Donna L.
- Published
- 2019
- Full Text
- View/download PDF
8. Single-cell transcriptomics of human T cells reveals tissue and activation signatures in health and disease
- Author
-
Szabo, Peter A., Levitin, Hanna Mendes, Miron, Michelle, Snyder, Mark E., Senda, Takashi, Yuan, Jinzhou, Cheng, Yim Ling, Bush, Erin C., Dogra, Pranay, Thapa, Puspa, Farber, Donna L., and Sims, Peter A.
- Published
- 2019
- Full Text
- View/download PDF
9. Effector CD8 T cells dedifferentiate into long-lived memory cells
- Author
-
Youngblood, Ben, Hale, Scott J., Kissick, Haydn T., Ahn, Eunseon, Xu, Xiaojin, Wieland, Andreas, Araki, Koichi, West, Erin E., Ghoneim, Hazem E., Fan, Yiping, Dogra, Pranay, Davis, Carl W., Konieczny, Bogumila T., Antia, Rustom, Cheng, Xiaodong, and Ahmed, Rafi
- Published
- 2017
- Full Text
- View/download PDF
10. Editorial: NK cells in viral immunology and immunotherapy.
- Author
-
Mulik, Sachin, Dogra, Pranay, and Jabrane-Ferrat, Nabila
- Subjects
KILLER cells ,IMMUNOLOGY ,IMMUNOTHERAPY ,IMMUNE response - Published
- 2023
- Full Text
- View/download PDF
11. SARS-CoV-2 infection generates tissue-localized immunological memory in humans.
- Author
-
Poon, Maya M.L., Rybkina, Ksenia, Kato, Yu, Kubota, Masaru, Matsumoto, Rei, Bloom, Nathaniel I., Zhang, Zeli, Hastie, Kathryn M., Grifoni, Alba, Weiskopf, Daniela, Wells, Steven B., Ural, Basak B., Lam, Nora, Szabo, Peter A., Dogra, Pranay, Lee, Yoon S., Gray, Joshua I., Bradley, Marissa C., Brusko, Maigan A., and Brusko, Todd M.
- Abstract
Adaptive immune responses to SARS-CoV-2 infection have been extensively characterized in blood; however, most functions of protective immunity must be accomplished in tissues. Here, we report from examination of SARS-CoV-2 seropositive organ donors (ages 10 to 74) that CD4+ T, CD8+ T, and B cell memory generated in response to infection is present in the bone marrow, spleen, lung, and multiple lymph nodes (LNs) for up to 6 months after infection. Lungs and lung-associated LNs were the most prevalent sites for SARS-CoV-2–specific memory T and B cells with significant correlations between circulating and tissue-resident memory T and B cells in all sites. We further identified SARS-CoV-2–specific germinal centers in the lung-associated LNs up to 6 months after infection. SARS-CoV-2–specific follicular helper T cells were also abundant in lung-associated LNs and lungs. Together, the results indicate local tissue coordination of cellular and humoral immune memory against SARS-CoV-2 for site-specific protection against future infectious challenges. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
12. Generation and persistence of human tissue-resident memory T cells in lung transplantation.
- Author
-
Snyder, Mark E., Finlayson, Michael O., Connors, Thomas J., Dogra, Pranay, Senda, Takashi, Bush, Erin, Carpenter, Dustin, Marboe, Charles, Benvenuto, Luke, Shah, Lori, Robbins, Hilary, Hook, Jaime L., Sykes, Megan, D'Ovidio, Frank, Bacchetta, Matthew, Sonett, Joshua R., Lederer, David J., Arcasoy, Selim, Sims, Peter A., and Farber, Donna L.
- Abstract
Transplanted memories: Parabiosis experiments in mice have shaped our understanding of the tissue retention properties of tissue-resident memory T cells (T
RM ). By studying donor and recipient T cells in transplanted lungs, Snyder et al. have provided a rare glimpse into the generation and maintenance of human TRM . Whereas donor T cells were barely detectable in blood within 10 weeks after transplantation, donor TRM were abundant and persisted in transplanted lungs for more than a year. Recipient T cells infiltrating the lung gradually acquired TRM profiles over time as determined by analyses of T cells from bronchoalveolar lavages. In this 20-patient cohort, persistence of donor lung TRM correlated with improved clinical outcome, although further studies are needed to understand their role in graft retention. Tissue-resident memory T cells (TRM ) maintain immunity in diverse sites as determined in mouse models, whereas their establishment and role in human tissues have been difficult to assess. Here, we investigated human lung TRM generation, maintenance, and function in airway samples obtained longitudinally from human leukocyte antigen (HLA)–disparate lung transplant recipients, where donor and recipient T cells could be localized and tracked over time. Donor T cells persist specifically in the lungs (and not blood) of transplant recipients and express high levels of TRM signature markers including CD69, CD103, and CD49a, whereas lung-infiltrating recipient T cells gradually acquire TRM phenotypes over months in vivo. Single-cell transcriptome profiling of airway T cells reveals that donor T cells comprise two TRM -like subsets with varying levels of expression of TRM -associated genes, whereas recipient T cells comprised non-TRM and similar TRM -like subpopulations, suggesting de novo TRM generation. Transplant recipients exhibiting higher frequencies of persisting donor TRM experienced fewer adverse clinical events such as primary graft dysfunction and acute cellular rejection compared with recipients with low donor TRM persistence, suggesting that monitoring TRM dynamics could be clinically informative. Together, our results provide spatial and temporal insights into how human TRM develop, function, persist, and affect tissue integrity within the complexities of lung transplantation. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
13. The D-form of a novel heparan binding peptide decreases cytomegalovirus infection in vivo and in vitro.
- Author
-
Pitt, Elisabeth A., Dogra, Pranay, Patel, Ravi S., Williams, Angela, Wall, Jonathan S., and Sparer, Tim E.
- Subjects
- *
CYTOMEGALOVIRUS disease treatment , *IMMUNOSUPPRESSION , *INTELLECTUAL disabilities , *NEPHROTOXICOLOGY , *IN vivo studies - Abstract
Human cytomegalovirus (HCMV) infection in utero can lead to congenital sensory neural hearing loss and mental retardation. Reactivation or primary infection can increase the morbidity and mortality in immune suppressed transplant recipients and AIDS patients. The current standard of care for HCMV disease is nucleoside analogs, which can be nephrotoxic. In addition resistance to current treatments is becoming increasingly common. In an effort to develop novel CMV treatments, we tested the effectiveness of the D-form of a novel heparan sulfate binding peptide, p5R D , at reducing infection of ganciclovir (GCV) resistant HCMVs in vitro and MCMV in vivo . HCMV infection was reduced by greater than 90% when cells were pretreated with p5R D . Because p5R D acts by a mechanism unrelated to those used by current antivirals, it was effective at reducing GCV resistant HCMVs by 85%. We show that p5R D is resistant to common proteases and serum inactivation, which likely contributed to its ability to significantly reduced infection of peritoneal exudate cells and viral loads in the spleen and the lungs in vivo . The ability of p5R D to reduce HCMV infectivity in vitro including GCV resistant HCMVs and MCMV infection in vivo suggests that this peptide could be a novel anti-CMV therapeutic. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
14. Generating long-lived CD8+ T-cell memory: Insights from epigenetic programs.
- Author
-
Dogra, Pranay, Ghoneim, Hazem E., Abdelsamed, Hossam A., and Youngblood, Ben
- Abstract
T-cell-based immunological memory has the potential to provide the host with life-long protection against pathogen reexposure and thus offers tremendous promise for the design of vaccines targeting chronic infections or cancer. In order to exploit this potential in the design of new vaccines, it is necessary to understand how and when memory T cells acquire their poised effector potential, and moreover, how they maintain these properties during homeostatic proliferation. To gain insight into the persistent nature of memory T-cell functions, investigators have turned their attention to epigenetic mechanisms. Recent efforts have revealed that many of the properties acquired among memory T cells are coupled to stable changes in DNA methylation and histone modifications. Furthermore, it has recently been reported that the delineating features among memory T cells subsets are also linked to distinct epigenetic events, such as permissive and repressive histone modifications and DNA methylation programs, providing exciting new hypotheses regarding their cellular ancestry. Here, we review recent studies focused on epigenetic programs acquired during effector and memory T-cell differentiation and discuss how these data may shed new light on the developmental path for generating long-lived CD8
+ T-cell memory. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
15. What We Have Learned from Animal Models of HCMV.
- Author
-
Dogra, Pranay and Sparer, Tim E.
- Abstract
Although human cytomegalovirus (HCMV) primary infection is generally asymptomatic, in immune-compromised patients HCMV increases morbidity and mortality. As a member of the betaherpesvirus family, in vivo studies of HCMV are limited due to its species specificity. CMVs from other species are often used as surrogates to express HCMV genes/proteins or used as models for inferring HCMV protein function in humans. Using innovative experiments, these animal models have answered important questions about CMV's life cycle, dissemination, pathogenesis, immune evasion, and host immune response. This chapter provides CMV biologists with an overview of the insights gained using these animal models. Subsequent chapters will provide details of the specifics of the experimental methods developed for each of the animal models discussed here. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
16. Novel Human Cytomegalovirus Viral Chemokines, vCXCL-ls, Display Functional Selectivity for Neutrophil Signaling and Function.
- Author
-
Heo, Jinho, Dogra, Pranay, Masi, Tom J., Pitt, Elisabeth A., de Kruij, Petra, Smit, Martine J., and Sparer, Tim E.
- Subjects
- *
HUMAN cytomegalovirus diseases , *CHEMOKINES , *NEUTROPHILS , *CELLULAR signal transduction , *G protein coupled receptors - Abstract
Human CMV (HCMV) uses members of the hematopoietic system including neutrophils for dissemination throughout the body. HCMV encodes a viral chemokine, vCXCL-1, that is postulated to attract neutrophils for dissemination within the host. The gene encoding vCXCL-1, UL146, is one of the most variable genes in the HCMV genome. Why HCMV has evolved this hypervariability and how this affects the virus' dissemination and pathogenesis is unknown. Because the vCXCL-1 hypervariability maps to important binding and activation domains, we hypothesized that vCXCL-1s differentially activate neutrophils, which could contribute to HCMV dissemination, pathogenesis, or both. To test whether these viral chemokines affect neutrophil function, we generated vCXCL-1 proteins from 11 different clades from clinical isolates from infants infected congenitally with HCMV. All vCXCL-1s were able to induce calcium flux at a concentration of 100 nM and integrin expression on human peripheral blood neutrophils, despite differences in affinity for the CXCR1 and CXCR2 receptors. In fact, their affinity for CXCR1 or CXCR2 did not correlate directly with chemotaxis, G protein-dependent and independent (β-arrestin-2) activation, or secondary chemokine (CCL22) expression. Our data suggest that vCXCL-1 polymorphisms affect the binding affinity, receptor usage, and differential peripheral blood neutrophil activation that could contribute to HCMV dissemination and pathogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
17. Novel Heparan Sulfate-Binding Peptides for Blocking Herpesvirus Entry.
- Author
-
Dogra, Pranay, Martin, Emily B., Williams, Angela, Richardson, Raphael L., Foster, James S., Hackenback, Nicole, Kennel, Stephen J., Sparer, Tim E., and Wall, Jonathan S.
- Subjects
- *
HEPARAN sulfate , *HERPESVIRUSES , *CYTOMEGALOVIRUS diseases , *DEAFNESS , *INTELLECTUAL disabilities , *IMMUNOSUPPRESSION - Abstract
Human cytomegalovirus (HCMV) infection can lead to congenital hearing loss and mental retardation. Upon immune suppression, reactivation of latent HCMV or primary infection increases morbidity in cancer, transplantation, and late stage AIDS patients. Current treatments include nucleoside analogues, which have significant toxicities limiting their usefulness. In this study we screened a panel of synthetic heparin-binding peptides for their ability to prevent CMV infection in vitro. A peptide designated, p5+14 exhibited ~ 90% reduction in murine CMV (MCMV) infection. Because negatively charged, cell-surface heparan sulfate proteoglycans (HSPGs), serve as the attachment receptor during the adsorption phase of the CMV infection cycle, we hypothesized that p5+14 effectively competes for CMV adsorption to the cell surface resulting in the reduction in infection. Positively charged Lys residues were required for peptide binding to cell-surface HSPGs and reducing viral infection. We show that this inhibition was not due to a direct neutralizing effect on the virus itself and that the peptide blocked adsorption of the virus. The peptide also inhibited infection of other herpesviruses: HCMV and herpes simplex virus 1 and 2 in vitro, demonstrating it has broad-spectrum antiviral activity. Therefore, this peptide may offer an adjunct therapy for the treatment of herpes viral infections and other viruses that use HSPGs for entry. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
18. Heterogeneity of human anti-viral immunity shaped by virus, tissue, age, and sex.
- Author
-
Poon, Maya M.L., Byington, Eve, Meng, Wenzhao, Kubota, Masaru, Matsumoto, Rei, Grifoni, Alba, Weiskopf, Daniela, Dogra, Pranay, Lam, Nora, Szabo, Peter A., Ural, Basak Burcu, Wells, Steven B., Rosenfeld, Aaron M., Brusko, Maigan A., Brusko, Todd M., Connors, Thomas J., Sette, Alessandro, Sims, Peter A., Luning Prak, Eline T., and Shen, Yufeng
- Abstract
The persistence of anti-viral immunity is essential for protection and exhibits profound heterogeneity across individuals. Here, we elucidate the factors that shape maintenance and function of anti-viral T cell immunity in the body by comprehensive profiling of virus-specific T cells across blood, lymphoid organs, and mucosal tissues of organ donors. We use flow cytometry, T cell receptor sequencing, single-cell transcriptomics, and cytokine analysis to profile virus-specific CD8
+ T cells recognizing the ubiquitous pathogens influenza and cytomegalovirus. Our results reveal that virus specificity determines overall magnitude, tissue distribution, differentiation, and clonal repertoire of virus-specific T cells. Age and sex influence T cell differentiation and dissemination in tissues, while T cell tissue residence and functionality are highly correlated with the site. Together, our results demonstrate how the covariates of virus, tissue, age, and sex impact the anti-viral immune response, which is important for targeting, monitoring, and predicting immune responses to existing and emerging viruses. [Display omitted] • CD8+ T cells specific for influenza and CMV localize in multiple tissue sites • Persistence of memory CD8+ T cell subsets is shaped by virus tropism and specificity • Regulation of functional responses to viral antigens is primarily tissue mediated • Age and sex influence T cell subset differentiation and tissue residency Through comprehensive cellular and molecular analysis of virus-specific T cells in circulation and across multiple lymphoid and mucosal tissues, Poon et al. elucidate how maintenance and function of the human anti-viral immune response against ubiquitous viruses influenza and cytomegalovirus are shaped by virus, tissue, age, and sex. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
19. Longitudinal profiling of respiratory and systemic immune responses reveals myeloid cell-driven lung inflammation in severe COVID-19.
- Author
-
Szabo, Peter A., Dogra, Pranay, Gray, Joshua I., Wells, Steven B., Connors, Thomas J., Weisberg, Stuart P., Krupska, Izabela, Matsumoto, Rei, Poon, Maya M.L., Idzikowski, Emma, Morris, Sinead E., Pasin, Chloé, Yates, Andrew J., Ku, Amy, Chait, Michael, Davis-Porada, Julia, Guo, Xinzheng V., Zhou, Jing, Steinle, Matthew, and Mackay, Sean
- Subjects
- *
COVID-19 , *PNEUMONIA , *IMMUNE response , *PATHOLOGY , *PULMONARY circulation - Abstract
Immune response dynamics in coronavirus disease 2019 (COVID-19) and their severe manifestations have largely been studied in circulation. Here, we examined the relationship between immune processes in the respiratory tract and circulation through longitudinal phenotypic, transcriptomic, and cytokine profiling of paired airway and blood samples from patients with severe COVID-19 relative to heathy controls. In COVID-19 airways, T cells exhibited activated, tissue-resident, and protective profiles; higher T cell frequencies correlated with survival and younger age. Myeloid cells in COVID-19 airways featured hyperinflammatory signatures, and higher frequencies of these cells correlated with mortality and older age. In COVID-19 blood, aberrant CD163+ monocytes predominated over conventional monocytes, and were found in corresponding airway samples and in damaged alveoli. High levels of myeloid chemoattractants in airways suggest recruitment of these cells through a CCL2-CCR2 chemokine axis. Our findings provide insights into immune processes driving COVID-19 lung pathology with therapeutic implications for targeting inflammation in the respiratory tract. [Display omitted] • Airways show localized immune responses correlating to age and outcome in COVID-19 • Airway T cells are activated and resident, while myeloid cells are hyperinflammatory • Aberrant CD163hi and HLA-DRlo monocytes predominate in COVID-19 blood • Monocytes infiltrate airways and lung alveoli potentially through a CCL2-CCR2 axis Through longitudinal profiling of paired airways and blood from patients with severe COVID-19, Szabo et al. reveal airway immune responses that correlate with age and outcome. They further identify coordinate roles for T and myeloid cells in the respiratory tract and circulation in perpetuating lung pathology and disease pathogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
20. Tissue Determinants of Human NK Cell Development, Function, and Residence.
- Author
-
Dogra, Pranay, Rancan, Chiara, Ma, Wenji, Toth, Marta, Senda, Takashi, Carpenter, Dustin J., Kubota, Masaru, Matsumoto, Rei, Thapa, Puspa, Szabo, Peter A., Li Poon, Maya Meimei, Li, Jacky, Arakawa-Hoyt, Janice, Shen, Yufeng, Fong, Lawrence, Lanier, Lewis L., and Farber, Donna L.
- Subjects
- *
KILLER cells , *BONE marrow , *MUCOUS membranes , *LYMPHOID tissue , *HOUSING , *CELL analysis , *BIOLOGICAL adaptation - Abstract
Immune responses in diverse tissue sites are critical for protective immunity and homeostasis. Here, we investigate how tissue localization regulates the development and function of human natural killer (NK) cells, innate lymphocytes important for anti-viral and tumor immunity. Integrating high-dimensional analysis of NK cells from blood, lymphoid organs, and mucosal tissue sites from 60 individuals, we identify tissue-specific patterns of NK cell subset distribution, maturation, and function maintained across age and between individuals. Mature and terminally differentiated NK cells with enhanced effector function predominate in blood, bone marrow, spleen, and lungs and exhibit shared transcriptional programs across sites. By contrast, precursor and immature NK cells with reduced effector capacity populate lymph nodes and intestines and exhibit tissue-resident signatures and site-specific adaptations. Together, our results reveal anatomic control of NK cell development and maintenance as tissue-resident populations, whereas mature, terminally differentiated subsets mediate immunosurveillance through diverse peripheral sites. • High-resolution map of human NK cells shows tissue-driven distribution across ages • Differentiated NK cells predominate in blood, bone marrow, spleen, and lungs • Tissue-resident NK cells exhibit specific adaptations in mucosal and lymphoid sites • Lymph nodes and intestines are reservoirs for precursor and immature NK cells A study of the distribution and function of natural killer cells across various human tissues reveals anatomic control of their development as well as populations that mediate immunosurveillance systemically. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
21. Tissue-Resident Memory T Cells Mediate Immune Homeostasis in the Human Pancreas through the PD-1/PD-L1 Pathway.
- Author
-
Weisberg, Stuart P., Carpenter, Dustin J., Chait, Michael, Dogra, Pranay, Gartrell-Corrado, Robyn D., Chen, Andrew X., Campbell, Sean, Liu, Wei, Saraf, Pooja, Snyder, Mark E., Kubota, Masaru, Danzl, Nichole M., Schrope, Beth A., Rabadan, Raul, Saenger, Yvonne, Chen, Xiaojuan, and Farber, Donna L.
- Abstract
Non-recirculating tissue-resident memory T cells (TRMs) are the predominant T cell subset in diverse tissue sites, where they mediate protective immune responses in situ. Here, we reveal a role for TRM in maintaining immune homeostasis in the human pancreas through interactions with resident macrophages and the PD-1/PD-L1 inhibitory pathway. Using tissues obtained from organ donors, we identify that pancreas T cells comprise CD8
+ PD-1hi TRMs, which are phenotypically, functionally, and transcriptionally distinct compared to TRMs in neighboring jejunum and lymph node sites. Pancreas TRMs cluster with resident macrophages throughout the exocrine areas; TRM effector functions are enhanced by macrophage-derived co-stimulation and attenuated by the PD-1/PD-L1 pathways. Conversely, in samples from chronic pancreatitis, TRMs exhibit reduced PD-1 expression and reduced interactions with macrophages. These findings suggest important roles for PD-1 and TRM-macrophage interactions in controlling tissue homeostasis and immune dysfunctions underlying inflammatory disease, with important implications for PD-1-based immunotherapies. • The human pancreas contains CD8+ TRMs exhibiting tissue-specific molecular signatures • Pancreas TRMs express high levels of PD-1 yet maintain strong effector function • During homeostasis, pancreas TRMs are regulated by PD-L1+ tissue macrophages • In chronic pancreatitis, TRM PD-1 levels and PD-L1+ macrophage density are reduced Non-recirculating tissue-resident memory T cells (TRMs) mediate immune responses in non-lymphoid tissues. Using a human organ donor tissue resource, Weisberg et al. reveal that PD-1hi pancreas TRMs are regulated by PD-L1+ macrophages during homeostasis. Comparison with chronic pancreatitis patient samples shows how pancreas TRM regulation is altered during inflammation. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
22. De Novo Epigenetic Programs Inhibit PD-1 Blockade-Mediated T Cell Rejuvenation.
- Author
-
Ghoneim, Hazem E., Moustaki, Ardiana, Abdelsamed, Hossam A., Dash, Pradyot, Dogra, Pranay, Awad, Walid, Thomas, Paul G., Youngblood, Ben, Fan, Yiping, Carter, Robert, and Neale, Geoff
- Subjects
- *
T cells , *REJUVENATION , *FATIGUE (Physiology) , *EPIGENETICS , *DNA methylation - Abstract
Summary Immune-checkpoint-blockade (ICB)-mediated rejuvenation of exhausted T cells has emerged as a promising approach for treating various cancers and chronic infections. However, T cells that become fully exhausted during prolonged antigen exposure remain refractory to ICB-mediated rejuvenation. We report that blocking de novo DNA methylation in activated CD8 T cells allows them to retain their effector functions despite chronic stimulation during a persistent viral infection. Whole-genome bisulfite sequencing of antigen-specific murine CD8 T cells at the effector and exhaustion stages of an immune response identified progressively acquired heritable de novo methylation programs that restrict T cell expansion and clonal diversity during PD-1 blockade treatment. Moreover, these exhaustion-associated DNA-methylation programs were acquired in tumor-infiltrating PD-1hi CD8 T cells, and approaches to reverse these programs improved T cell responses and tumor control during ICB. These data establish de novo DNA-methylation programming as a regulator of T cell exhaustion and barrier of ICB-mediated T cell rejuvenation. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
23. Distinct Localization, Transcriptional Profiles, and Functionality in Early Life Tonsil Regulatory T Cells.
- Author
-
Verma S, Bradley MC, Gray J, Dogra P, Caron DP, Maurrasse S, Grunstein E, Waldman E, Jang M, Pethe K, Farber DL, and Connors TJ
- Subjects
- Humans, Child, Adult, Child, Preschool, Female, Male, Forkhead Transcription Factors genetics, Forkhead Transcription Factors metabolism, Transcriptome immunology, Infant, Adolescent, Interleukin-10 immunology, CD8-Positive T-Lymphocytes immunology, Gene Expression Profiling, T-Lymphocytes, Regulatory immunology, Palatine Tonsil immunology, Palatine Tonsil cytology
- Abstract
CD4+ regulatory T cells (Tregs) are key orchestrators of the immune system, fostering the establishment of protective immunity while preventing deleterious responses. Infancy and childhood are crucial periods of rapid immunologic development, but how Tregs mediate immune responses at these earliest timepoints of human life is poorly understood. In this study, we compare blood and tissue (tonsil) Tregs across pediatric and adult subjects to investigate age-related differences in Treg biology. We observed increased FOXP3 expression and proportions of Tregs in tonsil compared with paired blood samples in children. Within tonsil, early life Tregs accumulated in extrafollicular regions with cellular interactions biased toward CD8+ T cells. Tonsil Tregs in both children and adults expressed transcriptional profiles enriched for lineage defining signatures and canonical functionality compared with blood, suggesting tissue as the primary site of Treg activity. Early life tonsil Tregs transcriptional profiles were further defined by pathways associated with activation, proliferation, and polyfunctionality. Observed differences in pediatric tonsil Treg transcriptional signatures were associated with phenotypic differences, high proliferative capacity, and robust production of IL-10 compared with adult Tregs. These results identify tissue as a major driver of Treg identity, provide new insights into developmental differences in Treg biology across the human lifespan, and demonstrate unique functional properties of early life Tregs., (Copyright © 2024 by The American Association of Immunologists, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
24. Immune and epithelial determinants of age-related risk and alveolar injury in fatal COVID-19.
- Author
-
Chait M, Yilmaz MM, Shakil S, Ku AW, Dogra P, Connors TJ, Szabo PA, Gray JI, Wells SB, Kubota M, Matsumoto R, Poon MM, Snyder ME, Baldwin MR, Sims PA, Saqi A, Farber DL, and Weisberg SP
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Alveolar Epithelial Cells pathology, Autopsy, Humans, Lung pathology, Middle Aged, Young Adult, Acute Lung Injury pathology, COVID-19
- Abstract
Respiratory failure in COVID-19 is characterized by widespread disruption of the lung's alveolar gas exchange interface. To elucidate determinants of alveolar lung damage, we performed epithelial and immune cell profiling in lungs from 24 COVID-19 autopsies and 43 uninfected organ donors ages 18-92 years. We found marked loss of type 2 alveolar epithelial (T2AE) cells and increased perialveolar lymphocyte cytotoxicity in all fatal COVID-19 cases, even at early stages before typical patterns of acute lung injury are histologically apparent. In lungs from uninfected organ donors, there was also progressive loss of T2AE cells with increasing age, which may increase susceptibility to COVID-19-mediated lung damage in older individuals. In the fatal COVID-19 cases, macrophage infiltration differed according to the histopathological pattern of lung injury. In cases with acute lung injury, we found accumulation of CD4+ macrophages that expressed distinctly high levels of T cell activation and costimulation genes and strongly correlated with increased extent of alveolar epithelial cell depletion and CD8+ T cell cytotoxicity. Together, our results show that T2AE cell deficiency may underlie age-related COVID-19 risk and initiate alveolar dysfunction shortly after infection, and we define immune cell mediators that may contribute to alveolar injury in distinct pathological stages of fatal COVID-19.
- Published
- 2022
- Full Text
- View/download PDF
25. Analysis of respiratory and systemic immune responses in COVID-19 reveals mechanisms of disease pathogenesis.
- Author
-
Szabo PA, Dogra P, Gray JI, Wells SB, Connors TJ, Weisberg SP, Krupska I, Matsumoto R, Poon MML, Idzikowski E, Morris SE, Pasin C, Yates AJ, Ku A, Chait M, Davis-Porada J, Zhou J, Steinle M, Mackay S, Saqi A, Baldwin M, Sims PA, and Farber DL
- Abstract
Immune responses to respiratory viruses like SARS-CoV-2 originate and function in the lung, yet assessments of human immunity are often limited to blood. Here, we conducted longitudinal, high-dimensional profiling of paired airway and blood samples from patients with severe COVID-19, revealing immune processes in the respiratory tract linked to disease pathogenesis. Survival from severe disease was associated with increased CD4
+ T cells and decreased monocyte/macrophage frequencies in the airway, but not in blood. Airway T cells and macrophages exhibited tissue-resident phenotypes and activation signatures, including high level expression and secretion of monocyte chemoattractants CCL2 and CCL3 by airway macrophages. By contrast, monocytes in blood expressed the CCL2-receptor CCR2 and aberrant CD163+ and immature phenotypes. Extensive accumulation of CD163+ monocyte/macrophages within alveolar spaces in COVID-19 lung autopsies suggested recruitment from circulation. Our findings provide evidence that COVID-19 pathogenesis is driven by respiratory immunity, and rationale for site-specific treatment and prevention strategies.- Published
- 2020
- Full Text
- View/download PDF
26. Stealth Killing by Uterine NK Cells for Tolerance and Tissue Homeostasis.
- Author
-
Dogra P and Farber DL
- Subjects
- Bacteria, Female, Homeostasis, Humans, Pregnancy, Uterus, Killer Cells, Natural, Trophoblasts
- Abstract
Human natural killer (NK) cells are critical for innate defense against pathogens through direct cytotoxicity of infected cells and are the predominant immune cell at the maternal-fetal interface. In this issue of Cell, Crespo et al. show that human NK cells in the decidual region of the uterus can clear a bacterial infection from the developing fetus by infusion of granulysin into placental trophoblast cells via nanotubes, thus removing the intracellular pathogen without damage to the placental cell. These findings reveal a mechanism for targeted immune protection of the developing fetus that maintains tolerance at the maternal-fetal interface., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
27. Antibody responses to SARS-CoV2 are distinct in children with MIS-C compared to adults with COVID-19.
- Author
-
Weisberg SP, Connors T, Zhu Y, Baldwin M, Lin WH, Wontakal S, Szabo PA, Wells SB, Dogra P, Gray JI, Idzikowski E, Bovier F, Davis-Porada J, Matsumoto R, Li Poon MM, Chait MP, Mathieu C, Horvat B, Decimo D, Bitan ZC, La Carpia F, Ferrara SA, Mace E, Milner J, Moscona A, Hod EA, Porotto M, and Farber DL
- Abstract
Clinical manifestations of COVID-19 caused by the novel coronavirus SARS-CoV-2 are associated with age. While children are largely spared from severe respiratory disease, they can present with a SARS-CoV-2-associated multisystem inflammatory syndrome (MIS-C) similar to Kawasaki's disease. Here, we show distinct antibody (Ab) responses in children with MIS-C compared to adults with severe COVID-19 causing acute respiratory distress syndrome (ARDS), and those who recovered from mild disease. There was a reduced breadth and specificity of anti-SARS-CoV-2-specific antibodies in MIS-C patients compared to the COVID patient groups; MIS-C predominantly generated IgG Abs specific for the Spike (S) protein but not for the nucleocapsid (N) protein, while both COVID-19 cohorts had anti-S IgG, IgM and IgA Abs, as well as anti-N IgG Abs. Moreover, MIS-C patients had reduced neutralizing activity compared to COVID-19 cohorts, indicating a reduced protective serological response. These results suggest a distinct infection course and immune response in children and adults who develop severe disease, with implications for optimizing treatments based on symptom and age.
- Published
- 2020
- Full Text
- View/download PDF
28. The Human Cytomegalovirus Chemokine vCXCL-1 Modulates Normal Dissemination Kinetics of Murine Cytomegalovirus In Vivo .
- Author
-
Jackson JW, Hancock TJ, LaPrade E, Dogra P, Gann ER, Masi TJ, Panchanathan R, Miller WE, Wilhelm SW, and Sparer TE
- Subjects
- Animals, Chemokine CXCL1 genetics, Host-Pathogen Interactions immunology, Humans, Kinetics, Mice, Mice, Inbred BALB C, Muromegalovirus pathogenicity, Neutrophils immunology, Receptors, Interleukin-8B genetics, Receptors, Interleukin-8B immunology, Virulence Factors immunology, Virus Replication, Chemokine CXCL1 immunology, Cytomegalovirus immunology, Cytomegalovirus Infections immunology, Muromegalovirus immunology, Neutrophils virology
- Abstract
Human cytomegalovirus (HCMV) is a betaherpesvirus that is a significant pathogen within newborn and immunocompromised populations. Morbidity associated with HCMV infection is the consequence of viral dissemination. HCMV has evolved to manipulate the host immune system to enhance viral dissemination and ensure long-term survival within the host. The immunomodulatory protein vCXCL-1, a viral chemokine functioning primarily through the CXCR2 chemokine receptor, is hypothesized to attract CXCR2
+ neutrophils to infection sites, aiding viral dissemination. Neutrophils harbor HCMV in vivo ; however, the interaction between vCXCL-1 and the neutrophil has not been evaluated in vivo Using the mouse model and mouse cytomegalovirus (MCMV) infection, we show that murine neutrophils harbor and transfer infectious MCMV and that virus replication initiates within this cell type. Utilizing recombinant MCMVs expressing vCXCL-1 from the HCMV strain (Toledo), we demonstrated that vCXCL-1 significantly enhances MCMV dissemination kinetics. Through cellular depletion experiments, we observe that neutrophils impact dissemination but that overall dissemination is largely neutrophil independent. This work adds neutrophils to the list of innate cells (i.e., dendritic and macrophages/monocytes) that contribute to MCMV dissemination but refutes the hypothesis that neutrophils are the primary cell responding to vCXCL-1. IMPORTANCE An adequate in vivo analysis of HCMV's viral chemokine vCXCL-1 has been lacking. Here we generate recombinant MCMVs expressing vCXCL-1 to study vCXCL-1 function in vivo using MCMV as a surrogate. We demonstrate that vCXCL-1 increases MCMV dissemination kinetics for both primary and secondary dissemination. Additionally, we provide evidence, that the murine neutrophil is largely a bystander in the mouse's response to vCXCL-1. We confirm the hypothesis that vCXCL-1 is a HCMV virulence factor. Infection of severely immunocompromised mice with MCMVs expressing vCXCL-1 was lethal in more than 50% of infected animals, while all animals infected with parental virus survived during a 12-day period. This work provides needed insights into vCXCL-1 function in vivo ., (Copyright © 2019 Jackson et al.)- Published
- 2019
- Full Text
- View/download PDF
29. Anticytomegalovirus Peptides Point to New Insights for CMV Entry Mechanisms and the Limitations of In Vitro Screenings.
- Author
-
Jackson JW, Hancock TJ, Dogra P, Patel R, Arav-Boger R, Williams AD, Kennel SJ, Wall JS, and Sparer TE
- Subjects
- Animals, Cells, Cultured, Cytomegalovirus Infections drug therapy, Disease Models, Animal, Fibroblasts virology, Heparitin Sulfate metabolism, Humans, Mice, Mice, Inbred BALB C, Muromegalovirus drug effects, Virus Internalization drug effects, Virus Replication drug effects, Antiviral Agents pharmacology, Cytomegalovirus drug effects, Cytomegalovirus physiology, Cytomegalovirus Infections prevention & control, Peptides pharmacology
- Abstract
Human cytomegalovirus (HCMV) is a ubiquitous betaherpesvirus that can cause severe disease following in utero exposure, during primary infection, or latent virus reactivation in immunocompromised populations. These complications lead to a 1- to 2-billion-dollar economic burden, making vaccine development and/or alternative treatments a high priority. Current treatments for HCMV include nucleoside analogues such as ganciclovir (GCV), foscarnet, and cidofovir. Recently, letermovir, a terminase complex inhibitor, was approved for prophylaxis after stem cell transplantation. These treatments have unwanted side effects, and HCMV is becoming resistant to them. Therefore, we sought to develop an alternative treatment that targets a different stage in viral infection. Currently, small antiviral peptides are being investigated as anti-influenza and anti-HIV treatments. We have developed heparan sulfate-binding peptides as tools for preventing CMV infections. These peptides are highly effective at stopping infection of fibroblasts with in vitro- derived HCMV and murine cytomegalovirus (MCMV). However, they do not prevent MCMV infection in vivo Interestingly, these peptides inhibit infectivity of in vivo- derived CMVs, albeit not as well as tissue culture-grown CMVs. We further demonstrate that this class of heparan sulfate-binding peptides is incapable of inhibiting MCMV cell-to-cell spread, which is independent of heparan sulfate usage. These data indicate that inhibition of CMV infection can be achieved using synthetic polybasic peptides, but cell-to-cell spread and in vivo -grown CMVs require further investigation to design appropriate anti-CMV peptides. IMPORTANCE In the absence of an effective vaccine to prevent HCMV infections, alternative interventions must be developed. Prevention of viral entry into susceptible cells is an attractive alternative strategy. Here we report that heparan sulfate-binding peptides effectively inhibit entry into fibroblasts of in vitro- derived CMVs and partially inhibit in vivo- derived CMVs. This includes the inhibition of urine-derived HCMV (uCMV), which is highly resistant to antibody neutralization. While these antiviral peptides are highly effective at inhibiting cell-free virus, they do not inhibit MCMV cell-to-cell spread. This underscores the need to understand the mechanism of cell-to-cell spread and differences between in vivo -derived versus in vitro- derived CMV entry to effectively prevent CMV's spread., (Copyright © 2019 Jackson et al.)
- Published
- 2019
- Full Text
- View/download PDF
30. SNP-mediated disruption of CTCF binding at the IFITM3 promoter is associated with risk of severe influenza in humans.
- Author
-
Allen EK, Randolph AG, Bhangale T, Dogra P, Ohlson M, Oshansky CM, Zamora AE, Shannon JP, Finkelstein D, Dressen A, DeVincenzo J, Caniza M, Youngblood B, Rosenberger CM, and Thomas PG
- Subjects
- Alleles, Blotting, Western, CCCTC-Binding Factor, CD8-Positive T-Lymphocytes immunology, DNA Methylation, Genetic Predisposition to Disease, Genotype, Humans, Influenza, Human immunology, Membrane Proteins immunology, Nasal Lavage Fluid cytology, Polymorphism, Single Nucleotide, Quantitative Trait Loci, RNA-Binding Proteins immunology, Severity of Illness Index, Influenza, Human genetics, Interferon Regulatory Factor-3 metabolism, Membrane Proteins genetics, Promoter Regions, Genetic genetics, RNA-Binding Proteins genetics, Repressor Proteins metabolism
- Abstract
Previous studies have reported associations of IFITM3 SNP rs12252 with severe influenza, but evidence of association and the mechanism by which risk is conferred remain controversial. We prioritized SNPs in IFITM3 on the basis of putative biological function and identified rs34481144 in the 5' UTR. We found evidence of a new association of rs34481144 with severe influenza in three influenza-infected cohorts characterized by different levels of influenza illness severity. We determined a role for rs34481144 as an expression quantitative trait locus (eQTL) for IFITM3, with the risk allele associated with lower mRNA expression. The risk allele was found to have decreased IRF3 binding and increased CTCF binding in promoter-binding assays, and risk allele carriage diminished transcriptional correlations among IFITM3-neighboring genes, indicative of CTCF boundary activity. Furthermore, the risk allele disrupts a CpG site that undergoes differential methylation in CD8
+ T cell subsets. Carriers of the risk allele had reduced numbers of CD8+ T cells in their airways during natural influenza infection, consistent with IFITM3 promoting accumulation of CD8+ T cells in airways and indicating that a critical function for IFITM3 may be to promote immune cell persistence at mucosal sites.Our study identifies a new regulator of IFITM3 expression that associates with CD8+ T cell levels in the airways and a spectrum of clinical outcomes.- Published
- 2017
- Full Text
- View/download PDF
31. Human memory CD8 T cell effector potential is epigenetically preserved during in vivo homeostasis.
- Author
-
Abdelsamed HA, Moustaki A, Fan Y, Dogra P, Ghoneim HE, Zebley CC, Triplett BM, Sekaly RP, and Youngblood B
- Subjects
- Adoptive Transfer, CD8-Positive T-Lymphocytes drug effects, Cell Differentiation drug effects, Cell Differentiation immunology, Cell Proliferation drug effects, Cellular Reprogramming drug effects, Cellular Reprogramming genetics, Cytokines pharmacology, DNA Methylation drug effects, DNA Methylation genetics, Genetic Loci, Genome, Human, Hematopoietic Stem Cell Transplantation, Hepatocyte Nuclear Factor 1-alpha metabolism, Homeostasis drug effects, Humans, Immunocompromised Host, Immunologic Memory drug effects, Lymphocyte Subsets drug effects, Lymphocyte Subsets immunology, Phenotype, Receptors, CCR7 metabolism, Tissue Donors, CD8-Positive T-Lymphocytes immunology, Epigenesis, Genetic drug effects, Homeostasis genetics, Homeostasis immunology, Immunologic Memory genetics
- Abstract
Antigen-independent homeostasis of memory CD8 T cells is vital for sustaining long-lived T cell-mediated immunity. In this study, we report that maintenance of human memory CD8 T cell effector potential during in vitro and in vivo homeostatic proliferation is coupled to preservation of acquired DNA methylation programs. Whole-genome bisulfite sequencing of primary human naive, short-lived effector memory (T
EM ), and longer-lived central memory (TCM ) and stem cell memory (TSCM ) CD8 T cells identified effector molecules with demethylated promoters and poised for expression. Effector-loci demethylation was heritably preserved during IL-7- and IL-15-mediated in vitro cell proliferation. Conversely, cytokine-driven proliferation of TCM and TSCM memory cells resulted in phenotypic conversion into TEM cells and was coupled to increased methylation of the CCR7 and Tcf7 loci. Furthermore, haploidentical donor memory CD8 T cells undergoing in vivo proliferation in lymphodepleted recipients also maintained their effector-associated demethylated status but acquired TEM -associated programs. These data demonstrate that effector-associated epigenetic programs are preserved during cytokine-driven subset interconversion of human memory CD8 T cells., (© 2017 Abdelsamed et al.)- Published
- 2017
- Full Text
- View/download PDF
32. A little cooperation helps murine cytomegalovirus (MCMV) go a long way: MCMV co-infection rescues a chemokine salivary gland defect.
- Author
-
Dogra P, Miller-Kittrell M, Pitt E, Jackson JW, Masi T, Copeland C, Wu S, Miller WE, and Sparer T
- Subjects
- Adaptive Immunity, Animals, Chemokines, CXC genetics, Cytomegalovirus Infections immunology, Cytomegalovirus Infections virology, Host-Pathogen Interactions, Immunity, Innate, Mice, Mice, SCID, Muromegalovirus genetics, Pan troglodytes, Salivary Glands immunology, Viral Proteins genetics, Chemokines, CXC immunology, Cytomegalovirus Infections veterinary, Muromegalovirus immunology, Salivary Glands virology, Viral Proteins immunology
- Abstract
Cytomegaloviruses (CMVs) produce chemokines (vCXCLs) that have both sequence and functional homology to host chemokines. Assessment of vCXCL-1's role in CMV infection is limited to in vitro and in silico analysis due to CMVs species specificity. In this study, we used the murine CMV (MCMV) mouse model to evaluate the function of vCXCL-1 in vivo. Recombinant MCMVs expressing chimpanzee CMV vCXCL-1 (vCXCL-1CCMV) or host chemokine, mCXCL1, underwent primary dissemination to the popliteal lymph node, spleen and lung similar to the parental MCMV. However, neither of the recombinants expressing chemokines was recovered from the salivary gland (SG) at any time post-infection although viral DNA was detected. This implies that the virus does not grow in the SG or the overexpressed chemokine induces an immune response that leads to suppressed growth. Pointing to immune suppression of virus replication, recombinant viruses were isolated from the SG following infection of immune-ablated mice [i.e. SCID (severe combined immunodeficiency), NSG (non-obese diabetic SCID gamma) or cyclophosphamide treated]. Depletion of neutrophils or NK cells does not rescue the recovery of chemokine-expressing recombinants in the SG. Surprisingly we found that co-infection of parental virus and chemokine-expressing virus leads to the recovery of the recombinants in the SG. We suggest that parental virus reduces the levels of chemokine expression leading to a decrease in inflammatory monocytes and subsequent SG growth. Therefore, aberrant expression of the chemokines induces cells of the innate and adaptive immune system that curtail the growth and dissemination of the recombinants in the SG.
- Published
- 2016
- Full Text
- View/download PDF
33. Generating long-lived CD8(+) T-cell memory: Insights from epigenetic programs.
- Author
-
Dogra P, Ghoneim HE, Abdelsamed HA, and Youngblood B
- Subjects
- Animals, CD8-Positive T-Lymphocytes cytology, Cell Differentiation genetics, Cell Differentiation immunology, Cellular Reprogramming genetics, Cellular Reprogramming immunology, DNA Methylation, Histones metabolism, Humans, Lymphocyte Activation genetics, Lymphocyte Activation immunology, Transcription, Genetic, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Epigenesis, Genetic, Gene Expression Regulation, Immunologic Memory genetics
- Abstract
T-cell-based immunological memory has the potential to provide the host with life-long protection against pathogen reexposure and thus offers tremendous promise for the design of vaccines targeting chronic infections or cancer. In order to exploit this potential in the design of new vaccines, it is necessary to understand how and when memory T cells acquire their poised effector potential, and moreover, how they maintain these properties during homeostatic proliferation. To gain insight into the persistent nature of memory T-cell functions, investigators have turned their attention to epigenetic mechanisms. Recent efforts have revealed that many of the properties acquired among memory T cells are coupled to stable changes in DNA methylation and histone modifications. Furthermore, it has recently been reported that the delineating features among memory T cells subsets are also linked to distinct epigenetic events, such as permissive and repressive histone modifications and DNA methylation programs, providing exciting new hypotheses regarding their cellular ancestry. Here, we review recent studies focused on epigenetic programs acquired during effector and memory T-cell differentiation and discuss how these data may shed new light on the developmental path for generating long-lived CD8(+) T-cell memory., (© 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2016
- Full Text
- View/download PDF
34. The inflammasome NLRP3 plays a protective role against a viral immunopathological lesion.
- Author
-
Gimenez F, Bhela S, Dogra P, Harvey L, Varanasi SK, Jaggi U, and Rouse BT
- Subjects
- Animals, Cornea immunology, Cornea pathology, Cornea virology, Female, Inflammation Mediators metabolism, Interleukin-18 metabolism, Interleukin-1beta metabolism, Keratitis virology, Lymph Nodes immunology, Lymph Nodes pathology, Mice, Inbred C57BL, NLR Family, Pyrin Domain-Containing 3 Protein deficiency, Neutrophils pathology, Th1 Cells immunology, Th17 Cells immunology, Herpesvirus 1, Human physiology, Inflammasomes metabolism, Keratitis immunology, Keratitis pathology, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Protective Agents metabolism
- Abstract
Herpes simplex 1 infection of the eye can cause blindness with lesions in the corneal stroma largely attributable to inflammatory events that include components of both adaptive and innate immunity. Several innate immune responses are triggered by herpes simplex 1, but it is unclear how such innate events relate to the subsequent development of stromal keratitis. In this study, we compared the outcome of herpes simplex 1 ocular infection in mice unable to express NLRP3 because of gene knockout (NLRP3(-/-)) to that of wild-type mice. The NLRP3(-/-) mice developed more-severe and earlier stromal keratitis lesions and had higher angiogenesis scores than did infected wild-type animals. In addition, NLRP3(-/-) mice generated an increased early immune response with heightened chemokines and cytokines, including interleukin-1β and interleukin-18, and elevated recruitment of neutrophils. Increased numbers of CD4(+) T cells were seen at later stages of the disease in NLRP3(-/-) animals. Reduction in neutrophils prevented early onset of the disease in NLRP3(-/-) animals and lowered levels of bioactive interleukin-1β but did not lower bioactive interleukin-18. In conclusion, our results indicate that NLRP3 has a regulatory and beneficial role in herpetic stromal keratitis pathogenesis., (© Society for Leukocyte Biology.)
- Published
- 2016
- Full Text
- View/download PDF
35. What we have learned from animal models of HCMV.
- Author
-
Dogra P and Sparer TE
- Subjects
- Animals, Cytomegalovirus immunology, Cytomegalovirus pathogenicity, Cytomegalovirus Infections genetics, Cytomegalovirus Infections virology, Disease Models, Animal, Humans, Cytomegalovirus genetics, Immune Evasion, Molecular Biology methods
- Abstract
Although human cytomegalovirus (HCMV) primary infection is generally asymptomatic, in immune-compromised patients HCMV increases morbidity and mortality. As a member of the betaherpesvirus family, in vivo studies of HCMV are limited due to its species specificity. CMVs from other species are often used as surrogates to express HCMV genes/proteins or used as models for inferring HCMV protein function in humans. Using innovative experiments, these animal models have answered important questions about CMV's life cycle, dissemination, pathogenesis, immune evasion, and host immune response. This chapter provides CMV biologists with an overview of the insights gained using these animal models. Subsequent chapters will provide details of the specifics of the experimental methods developed for each of the animal models discussed here.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.