Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where So-Yon Lim is active.

Publication


Featured researches published by So-Yon Lim.


Science Translational Medicine | 2011

Immune and Genetic Correlates of Vaccine Protection Against Mucosal Infection by SIV in Monkeys.

Norman L. Letvin; Srinivas S. Rao; David C. Montefiori; Michael S. Seaman; Yue Sun; So-Yon Lim; Wendy W. Yeh; Mohammed Asmal; Rebecca Gelman; Ling Shen; James B. Whitney; Cathal Seoighe; Miguel Lacerda; Sheila M. Keating; Philip J. Norris; Michael G. Hudgens; Peter B. Gilbert; Adam P. Buzby; Linh Mach; Jinrong Zhang; Harikrishnan Balachandran; George M. Shaw; Stephen D. Schmidt; John Paul Todd; Alan Dodson; John R. Mascola; Gary J. Nabel

A vaccine protecting monkeys against mucosal infection by simian immunodeficiency virus sheds light on immune and genetic correlates of protection. Unraveling Immune Correlates of Vaccine Protection Developing an effective vaccine against HIV-1, the virus that causes AIDS, has been a huge challenge that has stymied AIDS researchers for several decades. A key problem for HIV vaccine trials has been the lack of immune correlates that indicate which antibody and T cell responses in the vaccinees correlate directly with a protective effect. The only HIV vaccine trial to date that has shown a protective effect is the RV144 trial carried out in Thailand between 2003 and 2006, with the final results reported in 2009. In this trial of 16,400 Thai volunteers, those vaccinated with a prime-boost HIV vaccine showed a reduction in the rate of infection by HIV-1 of 31% compared to volunteers given a placebo. The protective effect was seen for up to 3 years after the initial vaccination, but the immune correlates of protection by this vaccine are still not known. In an effort to learn more about possible immune correlates of HIV vaccine protection, Letvin and colleagues used a prime/boost vaccine regimen in monkeys that was similar to that used in the RV144 trial. Monkeys were vaccinated with a plasmid DNA prime/recombinant adenovirus serotype 5 (rAd5) boost vaccine regimen and then were challenged with intrarectal doses of one of two isolates of the simian immunodeficiency virus (SIV) every week for 12 weeks. Although the vaccine had no impact on acquisition of the SIVmac251 isolate (which is tough for the monkey immune system to neutralize), the vaccine provided a 50% reduction in infection with the SIVsmE660 isolate (which more readily undergoes neutralization). The authors then examined a variety of immune responses in the protected vaccinated monkeys including cellular, antibody, and innate immune responses; they also examined whether protective host alleles were present in the protected animals. They found that low levels of neutralizing antibodies and a CD4+ T cell response against the HIV envelope (Env) protein correlated with the protective effect. In addition, monkeys that expressed two TRIM5 alleles that help to restrict SIV replication in host cells were protected by the vaccine, whereas monkeys expressing one TRIM5 allele that is permissive for SIV replication were not. This study begins to unravel the immune and genetic correlates of protection in nonhuman primates and highlights the need to scrutinize these types of correlates in future trials of HIV vaccines in human volunteers. The RV144 vaccine trial in Thailand demonstrated that an HIV vaccine could prevent infection in humans and highlights the importance of understanding protective immunity against HIV. We used a nonhuman primate model to define immune and genetic mechanisms of protection against mucosal infection by the simian immunodeficiency virus (SIV). A plasmid DNA prime/recombinant adenovirus serotype 5 (rAd5) boost vaccine regimen was evaluated for its ability to protect monkeys from infection by SIVmac251 or SIVsmE660 isolates after repeat intrarectal challenges. Although this prime-boost vaccine regimen failed to protect against SIVmac251 infection, 50% of vaccinated monkeys were protected from infection with SIVsmE660. Among SIVsmE660-infected animals, there was about a one-log reduction in peak plasma virus RNA in monkeys expressing the major histocompatibility complex class I allele Mamu-A*01, implicating cytotoxic T lymphocytes in the control of SIV replication once infection is established. Among Mamu-A*01–negative monkeys challenged with SIVsmE660, no CD8+ T cell response or innate immune response was associated with protection against virus acquisition. However, low levels of neutralizing antibodies and an envelope-specific CD4+ T cell response were associated with vaccine protection in these monkeys. Moreover, monkeys that expressed two TRIM5 alleles that restrict SIV replication were more likely to be protected from infection than monkeys that expressed at least one permissive TRIM5 allele. This study begins to elucidate the mechanisms of vaccine protection against immunodeficiency viruses and highlights the need to analyze these immune and genetic correlates of protection in future trials of HIV vaccine strategies.


PLOS Pathogens | 2010

TRIM5α Modulates Immunodeficiency Virus Control in Rhesus Monkeys

So-Yon Lim; Thomas Rogers; Tiffany Chan; James B. Whitney; Jonghwa Kim; Joseph Sodroski; Norman L. Letvin

The cytoplasmic TRIM5α proteins of certain mammalian lineages efficiently recognize the incoming capsids of particular retroviruses and potently restrict infection in a species-specific manner. Successful retroviruses have evolved capsids that are less efficiently recognized by the TRIM5α proteins of the natural hosts. To address whether TRIM5α contributes to the outcome of retroviral infection in a susceptible host species, we investigated the impact of TRIM5 polymorphisms in rhesus monkeys on the course of a simian immunodeficiency virus (SIV) infection. Full-length TRIM5α cDNAs were derived from each of 79 outbred monkeys and sequenced. Associations were explored between the expression of particular TRIM5 alleles and both the permissiveness of cells to SIV infection in vitro and clinical sequelae of SIV infection in vivo. Natural variation in the TRIM5α B30.2(SPRY) domain influenced the efficiency of SIVmac capsid binding and the in vitro susceptibility of cells from the monkeys to SIVmac infection. We also show the importance in vivo of the interaction of SIVmac with different allelic forms of TRIM5, demonstrating that particular alleles are associated with as much as 1.3 median log difference in set-point viral loads in SIVmac-infected rhesus monkeys. Moreover, these allelic forms of TRIM5 were associated with the extent of loss of central memory (CM) CD4+ T cells and the rate of progression to AIDS in the infected monkeys. These findings demonstrate a central role for TRIM5α in limiting the replication of an immunodeficiency virus infection in a primate host.


Nature Medicine | 2016

Zika viral dynamics and shedding in rhesus and cynomolgus macaques

Christa E. Osuna; So-Yon Lim; Claire Deleage; Bryan D. Griffin; Derek R. Stein; Lukas T. Schroeder; Robert W. Omange; Katharine Best; Ma Luo; Peter Hraber; Hanne Andersen-Elyard; Erwing Fabian Cardozo Ojeda; Scott Huang; Dana L. Vanlandingham; Stephen Higgs; Alan S. Perelson; Jacob D. Estes; David Safronetz; Mark G. Lewis; James B. Whitney

Infection with Zika virus has been associated with serious neurological complications and fetal abnormalities. However, the dynamics of viral infection, replication and shedding are poorly understood. Here we show that both rhesus and cynomolgus macaques are highly susceptible to infection by lineages of Zika virus that are closely related to, or are currently circulating in, the Americas. After subcutaneous viral inoculation, viral RNA was detected in blood plasma as early as 1 d after infection. Viral RNA was also detected in saliva, urine, cerebrospinal fluid (CSF) and semen, but transiently in vaginal secretions. Although viral RNA during primary infection was cleared from blood plasma and urine within 10 d, viral RNA was detectable in saliva and seminal fluids until the end of the study, 3 weeks after the resolution of viremia in the blood. The control of primary Zika virus infection in the blood was correlated with rapid innate and adaptive immune responses. We also identified Zika RNA in tissues, including the brain and male and female reproductive tissues, during early and late stages of infection. Re-infection of six animals 45 d after primary infection with a heterologous strain resulted in complete protection, which suggests that primary Zika virus infection elicits protective immunity. Early invasion of Zika virus into the nervous system of healthy animals and the extent and duration of shedding in saliva and semen underscore possible concern for additional neurologic complications and nonarthropod-mediated transmission in humans.


PLOS Pathogens | 2016

A Subset of Latency-Reversing Agents Expose HIV-Infected Resting CD4+ T-Cells to Recognition by Cytotoxic T-Lymphocytes.

R. Brad Jones; Stefanie Mueller; Rachel O’Connor; Katherine Rimpel; Derek D. Sloan; Dan Karel; Hing C. Wong; Emily K. Jeng; Allison S. Thomas; James B. Whitney; So-Yon Lim; Colin Kovacs; Erika Benko; Sara Karandish; Szu-han Huang; Maria J. Buzon; Mathias Lichterfeld; Alivelu Irrinki; Jeffrey P. Murry; Angela Tsai; Helen Yu; Romas Geleziunas; Alicja Trocha; Mario A. Ostrowski; Darrell J. Irvine; Bruce D. Walker

Resting CD4+ T-cells harboring inducible HIV proviruses are a critical reservoir in antiretroviral therapy (ART)-treated subjects. These cells express little to no viral protein, and thus neither die by viral cytopathic effects, nor are efficiently cleared by immune effectors. Elimination of this reservoir is theoretically possible by combining latency-reversing agents (LRAs) with immune effectors, such as CD8+ T-cells. However, the relative efficacy of different LRAs in sensitizing latently-infected cells for recognition by HIV-specific CD8+ T-cells has not been determined. To address this, we developed an assay that utilizes HIV-specific CD8+ T-cell clones as biosensors for HIV antigen expression. By testing multiple CD8+ T-cell clones against a primary cell model of HIV latency, we identified several single agents that primed latently-infected cells for CD8+ T-cell recognition, including IL-2, IL-15, two IL-15 superagonists (IL-15SA and ALT-803), prostratin, and the TLR-2 ligand Pam3CSK4. In contrast, we did not observe CD8+ T-cell recognition of target cells following treatment with histone deacetylase inhibitors or with hexamethylene bisacetamide (HMBA). In further experiments we demonstrate that a clinically achievable concentration of the IL-15 superagonist ‘ALT-803’, an agent presently in clinical trials for solid and hematological tumors, primes the natural ex vivo reservoir for CD8+ T-cell recognition. Thus, our results establish a novel experimental approach for comparative evaluation of LRAs, and highlight ALT-803 as an LRA with the potential to synergize with CD8+ T-cells in HIV eradication strategies.


Journal of Virology | 2011

The TRIM5 Gene Modulates Penile Mucosal Acquisition of Simian Immunodeficiency Virus in Rhesus Monkeys

Wendy W. Yeh; Srinivas S. Rao; So-Yon Lim; Jinrong Zhang; Peter Hraber; Laura M. Brassard; Corinne Luedemann; John Paul Todd; Alan Dodson; Ling Shen; Adam P. Buzby; James B. Whitney; Bette T. Korber; Gary J. Nabel; John R. Mascola; Norman L. Letvin

ABSTRACT There is considerable variability in host susceptibility to human immunodeficiency virus type 1 (HIV-1) infection, but the host genetic determinants of that variability are not well understood. In addition to serving as a block for cross-species retroviral infection, TRIM5 was recently shown to play a central role in limiting primate immunodeficiency virus replication. We hypothesized that TRIM5 may also contribute to susceptibility to mucosal acquisition of simian immunodeficiency virus (SIV) in rhesus monkeys. We explored this hypothesis by establishing 3 cohorts of Indian-origin rhesus monkeys with different TRIM5 genotypes: homozygous restrictive, heterozygous permissive, and homozygous permissive. We then evaluated the effect of TRIM5 genotype on the penile transmission of SIVsmE660. We observed a significant effect of TRIM5 genotype on mucosal SIVsmE660 acquisition in that no SIV transmission occurred in monkeys with only restrictive TRIM5 alleles. In contrast, systemic SIV infections were initiated after preputial pocket exposures in monkeys that had at least one permissive TRIM5 allele. These data demonstrate that host genetic factors can play a critical role in restricting mucosal transmission of a primate immunodeficiency virus. In addition, we used our understanding of TRIM5 to establish a novel nonhuman primate penile transmission model for AIDS mucosal pathogenesis and vaccine research.


PLOS Genetics | 2010

Contributions of Mamu-A*01 Status and TRIM5 Allele Expression, But Not CCL3L Copy Number Variation, to the Control of SIVmac251 Replication in Indian-Origin Rhesus Monkeys

So-Yon Lim; Tiffany Chan; Rebecca Gelman; James B. Whitney; Kara L. O'Brien; Dan H. Barouch; David B. Goldstein; Barton F. Haynes; Norman L. Letvin

CCL3 is a ligand for the HIV-1 co-receptor CCR5. There have recently been conflicting reports in the literature concerning whether CCL3-like gene (CCL3L) copy number variation (CNV) is associated with resistance to HIV-1 acquisition and with both viral load and disease progression following infection with HIV-1. An association has also been reported between CCL3L CNV and clinical sequelae of the simian immunodeficiency virus (SIV) infection in vivo in rhesus monkeys. The present study was initiated to explore the possibility of an association of CCL3L CNV with the control of virus replication and AIDS progression in a carefully defined cohort of SIVmac251-infected, Indian-origin rhesus monkeys. Although we demonstrated extensive variation in copy number of CCL3L in this cohort of monkeys, CCL3L CNV was not significantly associated with either peak or set-point plasma SIV RNA levels in these monkeys when MHC class I allele Mamu-A*01 was included in the models or progression to AIDS in these monkeys. With 66 monkeys in the study, there was adequate power for these tests if the correlation of CCL3L and either peak or set-point plasma SIV RNA levels was 0.34 or 0.36, respectively. These findings call into question the premise that CCL3L CNV is important in HIV/SIV pathogenesis.


Journal of Immune Based Therapies and Vaccines | 2006

Phytol-based novel adjuvants in vaccine formulation: 1. assessment of safety and efficacy during stimulation of humoral and cell-mediated immune responses

So-Yon Lim; Matt Meyer; Richard A. Kjonaas; Swapan K Ghosh

This is a correction article.


PLOS Pathogens | 2011

Association of Activating KIR Copy Number Variation of NK Cells with Containment of SIV Replication in Rhesus Monkeys

Ina Hellmann; So-Yon Lim; Rebecca Gelman; Norman L. Letvin

While the contribution of CD8+ cytotoxic T lymphocytes to early containment of HIV-1 spread is well established, a role for NK cells in controlling HIV-1 replication during primary infection has been uncertain. The highly polymorphic family of KIR molecules expressed on NK cells can inhibit or activate these effector cells and might therefore modulate their activity against HIV-1-infected cells. In the present study, we investigated copy number variation in KIR3DH loci encoding the only activating KIR receptor family in rhesus monkeys and its effect on simian immunodeficiency virus (SIV) replication during primary infection in rhesus monkeys. We observed an association between copy numbers of KIR3DH genes and control of SIV replication in Mamu-A*01– rhesus monkeys that express restrictive TRIM5 alleles. These findings provide further evidence for an association between NK cells and the early containment of SIV replication, and underscore the potential importance of activating KIRs in stimulating NK cell responses to control SIV spread.


Journal of Immune Based Therapies and Vaccines | 2006

Phytol-based novel adjuvants in vaccine formulation: 2. assessment of efficacy in the induction of protective immune responses to lethal bacterial infections in mice

So-Yon Lim; Adam Bauermeister; Richard A. Kjonaas; Swapan K. Ghosh

BackgroundAdjuvants are known to significantly enhance vaccine efficacy. However, commercial adjuvants often have limited use because of toxicity in humans. The objective of this study was to determine the comparative effectiveness of a diterpene alcohol, phytol and its hydrogenated derivative PHIS-01, relative to incomplete Freunds adjuvant (IFA), a commonly used adjuvant in augmenting protective immunity in mice against E. coli and S. aureus, and in terms of inflammatory cytokines.MethodsVaccines, consisting of heat-attenuated E. coli or S. aureus and either of the two phytol-based adjuvants or IFA, were tested in female BALB/c mice. The vaccines were administered intraperitoneally at 10-day intervals. The efficacy of the phytol and PHIS-01, as compared to IFA, was assessed by ELISA in terms of anti-bacterial antibody and inflammatory cytokines. We also examined the ability of the vaccines to induce specific protective immunity by challenging mice with different doses of live bacteria.Results and discussionIFA, phytol, and PHIS-01 were equally efficient in evoking anti-E. coli antibody response and in providing protective immunity against live E. coli challenges. In contrast, the antibody response to S. aureus was significant when PHIS-01 was used as the adjuvant. However, in terms of the ability to induce protective immunity, phytol was most effective against S. aureus. Moreover, during challenges with live E. coli and S. aureus immune mice produced much less IL-6, the mediators of fatal septic shock syndromes.ConclusionOur results show that vaccine formulations containing phytol and PHIS-01 as adjuvants confer a robust and protective immunity against both Gram-negative and Gram-positive bacteria without inducing adverse inflammatory cytokine due to IL-6.


Journal of Virology | 2015

Generation and Evaluation of Clade C Simian-Human Immunodeficiency Virus Challenge Stocks

Hui-Wen Chang; Lawrence J. Tartaglia; James B. Whitney; So-Yon Lim; Srisowmya Sanisetty; Christy L. Lavine; Michael S. Seaman; Cecelia Rademeyer; Carolyn Williamson; Katharine Ellingson-Strouss; Leonidas Stamatatos; James G. Kublin; Dan H. Barouch

ABSTRACT The development of a panel of mucosally transmissible simian-human immunodeficiency virus (SHIV) challenge stocks from multiple virus clades would facilitate preclinical evaluation of candidate HIV-1 vaccines and therapeutics. The majority of SHIV stocks that have been generated to date have been derived from clade B HIV-1 env sequences from viruses isolated during chronic infection and typically required serial animal-to-animal adaptation for establishing mucosal transmissibility and pathogenicity. To capture essential features of mucosal transmission of clade C viruses, we produced a series of SHIVs with early clade C HIV-1 env sequences from acutely HIV-1-infected individuals from South Africa. SHIV-327c and SHIV-327cRM expressed env sequences that were 99.7 to 100% identical to the original HIV-1 isolate and did not require in vivo passaging for mucosal infectivity. These challenge stocks infected rhesus monkeys efficiently by both intrarectal and intravaginal routes, replicated to high levels during acute infection, and established chronic setpoint viremia in 13 of 17 (76%) infected animals. The SHIV-327cRM challenge stock was also titrated for both single, high-dose intrarectal challenges and repetitive, low-dose intrarectal challenges in rhesus monkeys. These SHIV challenge stocks should facilitate the preclinical evaluation of vaccines and other interventions aimed at preventing clade C HIV-1 infection. IMPORTANCE We describe the development of two related clade C SHIV challenge stocks. These challenge stocks should prove useful for preclinical testing of vaccines and other interventions aimed at preventing clade C HIV-1 infection.

Collaboration


Dive into the So-Yon Lim's collaboration.

Top Co-Authors

Avatar

James B. Whitney

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Christa E. Osuna

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Norman L. Letvin

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peter Hraber

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael S. Seaman

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alan S. Perelson

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dan H. Barouch

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge