8 results on '"Rafick-Pierre Sekaly"'
Search Results
2. Interleukin-10 contributes to reservoir establishment and persistence in SIV-infected macaques treated with antiretroviral therapy
- Author
-
Justin Harper, Susan P. Ribeiro, Chi Ngai Chan, Malika Aid, Claire Deleage, Luca Micci, Maria Pino, Barbara Cervasi, Gopalan Raghunathan, Eric Rimmer, Gulesi Ayanoglu, Guoxin Wu, Neeta Shenvi, Richard J.O. Barnard, Gregory Q. Del Prete, Kathleen Busman-Sahay, Guido Silvestri, Deanna A. Kulpa, Steven E. Bosinger, Kirk A. Easley, Bonnie J. Howell, Dan Gorman, Daria J. Hazuda, Jacob D. Estes, Rafick-Pierre Sekaly, and Mirko Paiardini
- Subjects
AIDS/HIV ,Immunology ,Medicine - Abstract
Interleukin-10 (IL-10) is an immunosuppressive cytokine that signals through STAT3 to regulate T follicular helper (Tfh) cell differentiation and germinal center formation. In SIV-infected macaques, levels of IL-10 in plasma and lymph nodes (LNs) were induced by infection and not normalized with antiretroviral therapy (ART). During chronic infection, plasma IL-10 and transcriptomic signatures of IL-10 signaling were correlated with the cell-associated SIV-DNA content within LN CD4+ memory subsets, including Tfh cells, and predicted the frequency of CD4+ Tfh cells and their cell-associated SIV-DNA content during ART, respectively. In ART-treated rhesus macaques, cells harboring SIV-DNA by DNAscope were preferentially found in the LN B cell follicle in proximity to IL-10. Finally, we demonstrated that the in vivo neutralization of soluble IL-10 in ART-treated, SIV-infected macaques reduced B cell follicle maintenance and, by extension, LN memory CD4+ T cells, including Tfh cells and those expressing PD-1 and CTLA-4. Thus, these data support a role for IL-10 in maintaining a pool of target cells in lymphoid tissue that serve as a niche for viral persistence. Targeting IL-10 signaling to impair CD4+ T cell survival and improve antiviral immune responses may represent a novel approach to limit viral persistence in ART-suppressed people living with HIV.
- Published
- 2022
- Full Text
- View/download PDF
3. Lymph node CXCR5+ NK cells associate with control of chronic SHIV infection
- Author
-
Sheikh Abdul Rahman, James M. Billingsley, Ashish Arunkumar Sharma, Tiffany M. Styles, Sakthivel Govindaraj, Uma Shanmugasundaram, Hemalatha Babu, Susan Pereira Riberio, Syed A. Ali, Gregory K. Tharp, Chris Ibegbu, Stephen N. Waggoner, R. Paul Johnson, Rafick-Pierre Sekaly, Francois Villinger, Steve E. Bosinger, Rama Rao Amara, and Vijayakumar Velu
- Subjects
AIDS/HIV ,Immunology ,Medicine - Abstract
The persistence of virally infected cells as reservoirs despite effective antiretroviral therapy is a major barrier to an HIV/SIV cure. These reservoirs are predominately contained within cells present in the B cell follicles (BCFs) of secondary lymphoid tissues, a site that is characteristically difficult for most cytolytic antiviral effector cells to penetrate. Here, we identified a population of NK cells in macaque lymph nodes that expressed BCF-homing receptor CXCR5 and accumulated within BCFs during chronic SHIV infection. These CXCR5+ follicular NK cells exhibited an activated phenotype coupled with heightened effector functions and a unique transcriptome characterized by elevated expression of cytolytic mediators (e.g., perforin and granzymes, LAMP-1). CXCR5+ NK cells exhibited high expression of FcγRIIa and FcγRIIIa, suggesting a potential for elevated antibody-dependent effector functionality. Consistently, accumulation of CXCR5+ NK cells showed a strong inverse association with plasma viral load and the frequency of germinal center follicular Th cells that comprise a significant fraction of the viral reservoir. Moreover, CXCR5+ NK cells showed increased expression of transcripts associated with IL-12 and IL-15 signaling compared with the CXCR5– subset. Indeed, in vitro treatment with IL-12 and IL-15 enhanced the proliferation of CXCR5+ granzyme B+ NK cells. Our findings suggest that follicular homing NK cells might be important in immune control of chronic SHIV infection, and this may have important implications for HIV cure strategies.
- Published
- 2022
- Full Text
- View/download PDF
4. IFN-α blockade during ART-treated SIV infection lowers tissue vDNA, rescues immune function, and improves overall health
- Author
-
Louise A. Swainson, Ashish Arunkumar Sharma, Khader Ghneim, Susan Pereira Ribeiro, Peter Wilkinson, Richard M. Dunham, Rebecca G. Albright, Samson Wong, Jacob D. Estes, Michael Piatak, Steven G. Deeks, Peter W. Hunt, Rafick-Pierre Sekaly, and Joseph M. McCune
- Subjects
AIDS/HIV ,Immunology ,Medicine - Abstract
Type I IFNs (TI-IFNs) drive immune effector functions during acute viral infections and regulate cell cycling and systemic metabolism. That said, chronic TI-IFN signaling in the context of HIV infection treated with antiretroviral therapy (ART) also facilitates viral persistence, in part by promoting immunosuppressive responses and CD8+ T cell exhaustion. To determine whether inhibition of IFN-α might provide benefit in the setting of chronic, ART-treated SIV infection of rhesus macaques, we administered an anti–IFN-α antibody followed by an analytical treatment interruption (ATI). IFN-α blockade was well-tolerated and associated with lower expression of TI-IFN–inducible genes (including those that are antiviral) and reduced tissue viral DNA (vDNA). The reduction in vDNA was further accompanied by higher innate proinflammatory plasma cytokines, expression of monocyte activation genes, IL-12–induced effector CD8+ T cell genes, increased heme/metabolic activity, and lower plasma TGF-β levels. Upon ATI, SIV-infected, ART-suppressed nonhuman primates treated with anti–IFN-α displayed lower levels of weight loss and improved erythroid function relative to untreated controls. Overall, these data demonstrated that IFN-α blockade during ART-treated SIV infection was safe and associated with the induction of immune/erythroid pathways that reduced viral persistence during ART while mitigating the weight loss and anemia that typically ensue after ART interruption.
- Published
- 2022
- Full Text
- View/download PDF
5. The interferon paradox: can inhibiting an antiviral mechanism advance an HIV cure?
- Author
-
Pamela M. Odorizzi, Rafick-Pierre Sekaly, and Steven G. Deeks
- Subjects
0301 basic medicine ,Viral rebound ,Immunology ,Human immunodeficiency virus (HIV) ,HIV Infections ,medicine.disease_cause ,Antiviral mechanism ,Medical and Health Sciences ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Interferon ,medicine ,Animals ,Humans ,business.industry ,Animal ,Evaluation of treatments and therapeutic interventions ,General Medicine ,Human Fetal Tissue ,Blockade ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,Good Health and Well Being ,Anti-Retroviral Agents ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,6.1 Pharmaceuticals ,Humanized mouse ,Disease Models ,Interferon Type I ,Commentary ,HIV-1 ,HIV/AIDS ,Development of treatments and therapeutic interventions ,business ,Infection ,Interferon type I ,medicine.drug - Abstract
While antiretroviral therapy (ART) has improved the quality of life and increased the life span of many HIV-infected individuals, this therapeutic strategy has several limitations, including a lack of efficacy in fully restoring immune function and a requirement for life-long treatment. Two studies in this issue of the JCI use a humanized mouse model and demonstrate that type I interferon (IFN) is induced early during HIV infection and that type I IFN-associated gene signatures persist, even during ART. Importantly, blockade of type I IFN improved immune function, reduced the HIV reservoir, and caused a delay in viral rebound after ART interruption. Together, these two studies support further evaluation of IFN blockade as a supplement to ART.
- Published
- 2016
6. Immune activation alters cellular and humoral responses to yellow fever 17D vaccine
- Author
-
Sharon Isern, Rafael Cubas, Josephine Birungi, Lydie Trautmann, Josephine H. Cox, C. Schmidt, Jill Gillmour, Divya Srinivasan, Pearline Ngauv, Enoch Muyanja, Elias K. Haddad, Rafick Pierre Sekaly, Guido Silvestri, Jakub Kopycinski, Erika Castro, Benton Lawson, Pontiano Kaleebu, Giuseppe Pantaleo, Dawne K. Rowe, Scott F. Michael, Glenda Canderan, Noah Kiwanuka, Thomas H. Vanderford, Aloysius Ssemaganda, Michaela J. Smith, Joel Singer, Amanda S. Graham, Helene Perrin, Patricia E. Fast, Denis Gaucher, and Annet Nanvubya
- Subjects
Adult ,Male ,T cell ,Immunization, Secondary ,Yellow fever vaccine ,Adaptive Immunity ,CD8-Positive T-Lymphocytes ,Antibodies, Viral ,Lymphocyte Activation ,Vaccines, Attenuated ,Virus Replication ,Monocytes ,Cohort Studies ,Young Adult ,Immune system ,Immunity ,Yellow Fever ,Medicine ,Humans ,Uganda ,B cell ,B-Lymphocytes ,Immunity, Cellular ,business.industry ,Yellow Fever Vaccine ,General Medicine ,Middle Aged ,Acquired immune system ,Virology ,Antibodies, Neutralizing ,Immunity, Innate ,Immunity, Humoral ,Vaccination ,medicine.anatomical_structure ,Immunology ,Female ,Clinical Medicine ,Yellow fever virus ,Corrigendum ,business ,CD8 ,Switzerland ,medicine.drug - Abstract
Background. Defining the parameters that modulate vaccine responses in African populations will be imperative to design effective vaccines for protection against HIV, malaria, tuberculosis, and dengue virus infections. This study aimed to evaluate the contribution of the patient-specific immune microenvironment to the response to the licensed yellow fever vaccine 17D (YF-17D) in an African cohort. Methods. We compared responses to YF-17D in 50 volunteers in Entebbe, Uganda, and 50 volunteers in Lausanne, Switzerland. We measured the CD8+ T cell and B cell responses induced by YF-17D and correlated them with immune parameters analyzed by flow cytometry prior to vaccination. Results. We showed that YF-17D–induced CD8+ T cell and B cell responses were substantially lower in immunized individuals from Entebbe compared with immunized individuals from Lausanne. The impaired vaccine response in the Entebbe cohort associated with reduced YF-17D replication. Prior to vaccination, we observed higher frequencies of exhausted and activated NK cells, differentiated T and B cell subsets and proinflammatory monocytes, suggesting an activated immune microenvironment in the Entebbe volunteers. Interestingly, activation of CD8+ T cells and B cells as well as proinflammatory monocytes at baseline negatively correlated with YF-17D–neutralizing antibody titers after vaccination. Additionally, memory T and B cell responses in preimmunized volunteers exhibited reduced persistence in the Entebbe cohort but were boosted by a second vaccination. Conclusion. Together, these results demonstrate that an activated immune microenvironment prior to vaccination impedes efficacy of the YF-17D vaccine in an African cohort and suggest that vaccine regimens may need to be boosted in African populations to achieve efficient immunity. Trial registration. Registration is not required for observational studies. Funding. This study was funded by Canada’s Global Health Research Initiative, Defense Threat Reduction Agency, National Institute of Allergy and Infectious Diseases, Bill & Melinda Gates Foundation, and United States Agency for International Development.
- Published
- 2014
7. Loss of memory B cells during chronic HIV infection is driven by Foxo3a- and TRAIL-mediated apoptosis
- Author
-
Cécile Tremblay, Mohamed Rachid Boulassel, Elias K. Haddad, Rafael Cubas, Julien van Grevenynghe, Francesco A. Procopio, Alessandra Noto, Abdelali Filali-Mouhim, Zhong He, Franck P. Dupuy, Elias A. Said, Nicolas Chomont, Rafick Pierre Sekaly, Jean-Pierre Routy, Robert S. Balderas, Yoav Peretz, and Sandrina DaFonseca
- Subjects
Apoptosis ,business.industry ,Clinical investigation ,Immunology ,Human immunodeficiency virus (HIV) ,medicine ,General Medicine ,Corrigendum ,medicine.disease_cause ,business ,Virology - Published
- 2012
- Full Text
- View/download PDF
8. TCF-1 regulates HIV-specific CD8+ T cell expansion capacity
- Author
-
Rachel L. Rutishauser, Christian Deo T. Deguit, Joseph Hiatt, Franziska Blaeschke, Theodore L. Roth, Lynn Wang, Kyle A. Raymond, Carly E. Starke, Joseph C. Mudd, Wenxuan Chen, Carolyn Smullin, Rodrigo Matus-Nicodemos, Rebecca Hoh, Melissa Krone, Frederick M. Hecht, Christopher D. Pilcher, Jeffrey N. Martin, Richard A. Koup, Daniel C. Douek, Jason M. Brenchley, Rafick-Pierre Sékaly, Satish K. Pillai, Alexander Marson, Steven G. Deeks, Joseph M. McCune, and Peter W. Hunt
- Subjects
AIDS/HIV ,Immunology ,Medicine - Abstract
Although many HIV cure strategies seek to expand HIV-specific CD8+ T cells to control the virus, all are likely to fail if cellular exhaustion is not prevented. A loss in stem-like memory properties (i.e., the ability to proliferate and generate secondary effector cells) is a key feature of exhaustion; little is known, however, about how these properties are regulated in human virus–specific CD8+ T cells. We found that virus-specific CD8+ T cells from humans and nonhuman primates naturally controlling HIV/SIV infection express more of the transcription factor TCF-1 than noncontrollers. HIV-specific CD8+ T cell TCF-1 expression correlated with memory marker expression and expansion capacity and declined with antigenic stimulation. CRISPR-Cas9 editing of TCF-1 in human primary T cells demonstrated a direct role in regulating expansion capacity. Collectively, these data suggest that TCF-1 contributes to the regulation of the stem-like memory property of secondary expansion capacity of HIV-specific CD8+ T cells, and they provide a rationale for exploring the enhancement of this pathway in T cell–based therapeutic strategies for HIV.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.