Network


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

Hotspot


Dive into the research topics where Jianhong Cao is active.

Publication


Featured researches published by Jianhong Cao.


Journal of Experimental Medicine | 2007

Virus-specific CD8+ T cells accumulate near sensory nerve endings in genital skin during subclinical HSV-2 reactivation

Jia-Jia Zhu; David M. Koelle; Jianhong Cao; Julio A. Vázquez; Meei-Li Huang; Florian Hladik; Anna Wald; Lawrence Corey

Cytotoxic CD8+ T cells play a critical role in controlling herpes simplex virus (HSV) infection and reactivation. However, little is known about the spatiotemporal dynamics of CD8+ T cells during HSV lesion evolution or about their involvement in immune surveillance after lesion resolution. Using quantum dot–conjugated peptide–major histocompatibility complex multimers, we investigated the in vivo localization of HSV-2–specific CD8+ T cells in sequential biopsies of human genital skin during acute, resolving, and healed stages of HSV-2 reactivation. Our studies revealed that functionally active CD8+ T cells selectively infiltrated to the site of viral reactivation. After lesion healing in concert with complete reepithelialization and loss of HSV DNA from skin biopsies, HSV-2–specific CD8+ T cells persisted for more than two months at the dermal–epidermal junction, adjacent to peripheral nerve endings. In two out of the six sequentially studied individuals, HSV-2 DNA reappeared in clinically and histologically normal–appearing skin. Detection of viral DNA was accompanied by increased numbers of both HSV-specific and total CD8+ T cells in the dermis. These findings indicate that the frequency and clinical course of HSV-2 reactivation in humans is influenced by virus-specific CD8+ T cells that persist in peripheral mucosa and genital skin after resolution of herpes lesions.


Journal of Virology | 2006

Selection on the human immunodeficiency virus type 1 proteome following primary infection.

Yi Liu; John McNevin; Jianhong Cao; Hong Zhao; Indira Genowati; Kim Wong; Sherry McLaughlin; Matthew McSweyn; Kurt Diem; Claire E. Stevens; Janine Maenza; Hongxia He; David C. Nickle; Daniel Shriner; Sarah Holte; Ann C. Collier; Lawrence Corey; M. Juliana McElrath; James I. Mullins

ABSTRACT Typically during human immunodeficiency virus type 1 (HIV-1) infection, a nearly homogeneous viral population first emerges and then diversifies over time due to selective forces that are poorly understood. To identify these forces, we conducted an intensive longitudinal study of viral genetic changes and T-cell immunity in one subject at ≤17 time points during his first 3 years of infection, and in his infecting partner near the time of transmission. Autologous peptides covering amino acid sites inferred to be under positive selection were powerful for identifying HIV-1-specific cytotoxic-T-lymphocyte (CTL) epitopes. Positive selection and mutations resulting in escape from CTLs occurred across the viral proteome. We detected 25 CTL epitopes, including 14 previously unreported. Seven new epitopes mapped to the viral Env protein, emphasizing Env as a major target of CTLs. One-third of the selected sites were associated with epitopic mutational escapes from CTLs. Most of these resulted from replacement with amino acids found at low database frequency. Another one-third represented acquisition of amino acids found at high database frequency, suggesting potential reversions of CTL epitopic sites recognized by the immune system of the transmitting partner and mutation toward improved viral fitness in the absence of immune targeting within the recipient. A majority of the remaining selected sites occurred in the envelope protein and may have been subjected to humoral immune selection. Hence, a majority of the amino acids undergoing selection in this subject appeared to result from fitness-balanced CTL selection, confirming CTLs as a dominant selective force in HIV-1 infection.


Journal of Immunology | 2003

Evolution of CD8+ T Cell Immunity and Viral Escape Following Acute HIV-1 Infection

Jianhong Cao; John McNevin; Uma Malhotra; M. Juliana McElrath

Induction of HIV-1-specific CD8+ T cells during acute infection is associated with a decline in viremia. The role CD8+ effectors play in subsequently establishing viral set point remains unclear. To address this, we focused on two acutely infected patients with the same initial Tat-specific CD8+ response, analyzing their CD8+ T cell responses longitudinally in conjunction with viral load and sequence evolution. In one patient initiating treatment during acute infection, the frequencies of Tat-specific CD8+ T cells gradually diminished but persisted, and the Tat epitope sequence was unaltered. By contrast, in the second patient who declined treatment, the Tat-specific CD8+ T cells disappeared below detection, in conjunction with Gag-specific CD4+ T cell loss, as plasma viremia reached a set point. This coincided with the emergence of an escape variant within the Tat epitope and an additional Vpr epitope. New CD8+ T cell responses emerged but with no further associated decline in viremia. These findings indicate that, in the absence of treatment, the initial CD8+ T cell responses have the greatest impact on reducing viremia, and that later, continuously evolving responses are less efficient in further reducing viral load. The results also suggest that T cell help may contribute to the antiviral efficiency of the acute CD8+ T cell response.


Molecular and Cellular Biology | 1996

CODING SEQUENCE-DEPENDENT RIBOSOMAL ARREST AT TERMINATION OF TRANSLATION

Jianhong Cao; Adam P. Geballe

A remarkably high percentage of proto-oncogene, growth factor, cellular receptor, and viral transcript leaders contain short upstream open reading frames (uORFs), yet the significance and regulatory effects of these uORFs have not been well characterized. In the case of the human cytomegalovirus gpUL4 (gp48) transcript, the second of three uORFs (uORF2) inhibits translation of the downstream cistron by a process that depends on the uORF2 amino acid coding information. To investigate the mechanism underlying this unusual regulatory element, we adapted the toeprinting (or reverse transcriptase extension inhibition) assay for use in detecting positions of ribosomal stalling on gp48 transcripts. Using a cell-free translation system, we demonstrate that ribosomes arrest at the termination codon of uORF2 by a uORF2 coding sequence-dependent mechanism. Further, the sequence requirements for ribosomal stalling are the same as for inhibition of downstream translation. We also provide evidence for ribosomal stalling in vivo, on the natural viral mRNA. These data support the hypothesis that the inhibition of downstream translation results from uORF2 peptide-dependent ribosomal arrest at termination and suggest that translation termination may be a regulatory step in expression of some eukaryotic genes.


PLOS ONE | 2009

Fludarabine modulates immune response and extends in vivo survival of adoptively transferred CD8 T cells in patients with metastatic melanoma

Herschel Wallen; John A. Thompson; J. Zachary Reilly; Rebecca Rodmyre; Jianhong Cao; Cassian Yee

Background Adoptive T cell therapy involving the use of ex vivo generated antigen-specific cytotoxic T lymphocytes provides a promising approach to immunotherapy. It has become increasingly apparent that anti-tumor efficacy using adoptively transferred T cells is linked to their duration of in vivo persistence and can only be achieved when combined with some form of pre-infusion patient conditioning regimen. An optimal conditioning regimen that provides a positive benefit without serious toxicities has yet to be defined. We have established a unique clinical model that allows for evaluation of a given conditioning regimen on adoptively transferred T cells in humans. In this first-in-human study (FHCRC #1796), we evaluate the use of fludarabine, an FDA-approved reagent with predictable lymphodepleting kinetics and duration of action, as a conditioning regimen that promotes homeostatic upregulation of cytokines and growth signals contributing to in vivo T cell persistence. Methods/Findings We conducted a phase I study in patients with refractory metastatic melanoma. Patients received two infusions of a single tumor-reactive antigen-specific CTL clone expanded to 1010/m2; the first infusion was given without fludarabine conditioning, and the second CTL infusion was given after a course of fludarabine (25 mg/m2/day×5 days). This design permits intra-patient comparison of in vivo T cell persistence pre- and post-fludarabine. Nineteen CTL infusions were administered to ten patients. No serious toxicities were observed. Three of nine evaluable patients experienced minor response or stable disease for periods of 5.8–11.0 months with two additional patients demonstrating delayed disease stabilization. The median overall survival in this heavily pre-treated population was 9.7 months. Fludarabine led to a 2.9 fold improvement in the in vivo persistence of transferred CTL clones from a median of 4.5 days (range 0–38+) to 13.0 days (range 2–63+) (p<0.05). Fludarabine lymphodepletion increased plasma levels of the homeostatic cytokines IL-7 and IL-15. Surprisingly, fludarabine also increased the relative percentage of CD4+ T cells expressing the regulatory protein Foxp3. Conclusions/Significance Lymphodepletion with fludarabine enhances transferred T cell persistence but suggest that additional improvements to optimize T cell survival and address regulatory T cells are critical in providing anti-tumor efficacy. Trial Registration ClinicalTrials.gov NCT00317759


Molecular and Cellular Biology | 1996

Inhibition of Nascent-Peptide Release at Translation Termination

Jianhong Cao; Andadam P. Geballe

The transcript leader of the human cytomegalovirus (CMV) gpUL4 (gp48) gene contains a 22-codon upstream open reading frame (uORF2) that represses translation of the downstream cistron. Previous work demonstrated that ribosomes stall at the termination codon of uORF2 and, remarkably, that the coding information of uORF2 is required for both the translational repression and ribosomal stalling. We now provide evidence that the peptide product of uORF2 is synthesized and is retained in the ribosome in the form of a peptidyl-tRNA. Translation of the gp48 transcript leader in cell extracts produces the 2.4-kDa uORF2 peptide and a second product migrating with an apparent molecular mass of 20 kDa that represents the uORF2 peptide covalently linked to tRNA(Pro), the tRNA predicted to decode the carboxy-terminal codon of uORF2. The uORF2 peptidyl-tRNA is only detected after translation of RNAs containing uORF2 sequences that also inhibit downstream translation and cause ribosomal stalling. These data support a model in which the nascent uORF2 peptide blocks translation termination prior to hydrolysis of the peptidyl-tRNA bond. This blockade results in ribosomal stalling on the transcript leader which in turn impedes the access of ribosomes to the downstream cistron. This system illustrates that translation termination may be a critical step controlling expression of some eukaryotic genes.


Journal of Virology | 2003

Ontogeny and Specificities of Mucosal and Blood Human Immunodeficiency Virus Type 1-Specific CD8+ Cytotoxic T Lymphocytes

L. Musey; Y. Ding; Jianhong Cao; Jean M. Lee; C. Galloway; A. Yuen; Keith R. Jerome; M. J. McElrath

ABSTRACT Induction of adaptive immunity to human immunodeficiency virus type 1 (HIV-1) at the mucosal site of transmission is poorly understood but crucial in devising strategies to control and prevent infection. To gain further understanding of HIV-1-specific T-cell mucosal immunity, we established HIV-1-specific CD8+ cytotoxic T-lymphocyte (CTL) cell lines and clones from the blood, cervix, rectum, and semen of 12 HIV-1-infected individuals and compared their specificities, cytolytic function, and T-cell receptor (TCR) clonotypes. Blood and mucosal CD8+ CTL had common HIV-1 epitope specificities and major histocompatibility complex restriction patterns. Moreover, both systemic and mucosal CTL lysed targets with similar efficiency, primarily through the perforin-dependent pathway in in vitro studies. Sequence analysis of the TCRβ VDJ region revealed in some cases identical HIV-1-specific CTL clones in different compartments in the same HIV-1-infected individual. These results clearly establish that a subset of blood and mucosal HIV-1-specific CTL can have a common origin and can traffic between anatomically distinct compartments. Thus, these effectors can provide immune surveillance at the mucosa, where rapid responses are needed to contain HIV-1 infection.


Journal of Experimental Medicine | 2016

Combined IL-21–primed polyclonal CTL plus CTLA4 blockade controls refractory metastatic melanoma in a patient

Aude G. Chapuis; Sylvia Lee; John A. Thompson; Ilana M. Roberts; Kim Margolin; Shailender Bhatia; Heather L. Sloan; Ivy Lai; Felecia Wagener; Kendall C. Shibuya; Jianhong Cao; Jedd D. Wolchok; Philip D. Greenberg; Cassian Yee

Chapuis et al. demonstrate that the combination of adoptive cellular therapy with CTLA4 blockade induces long-term remission in a melanoma patient resistant to both modalities administered serially and individually.


Journal of Clinical Oncology | 2016

T-Cell Therapy Using Interleukin-21–Primed Cytotoxic T-Cell Lymphocytes Combined With Cytotoxic T-Cell Lymphocyte Antigen-4 Blockade Results in Long-Term Cell Persistence and Durable Tumor Regression

Aude G. Chapuis; Ilana M. Roberts; John A. Thompson; Kim Margolin; Shailender Bhatia; Sylvia Lee; Heather L. Sloan; Ivy Lai; Erik A. Farrar; Felecia Wagener; Kendall C. Shibuya; Jianhong Cao; Jedd D. Wolchok; Philip D. Greenberg; Cassian Yee

Purpose Peripheral blood-derived antigen-specific cytotoxic T cells (CTLs) provide a readily available source of effector cells that can be administered with minimal toxicity in an outpatient setting. In metastatic melanoma, this approach results in measurable albeit modest clinical responses in patients resistant to conventional therapy. We reasoned that concurrent cytotoxic T-cell lymphocyte antigen-4 (CTLA-4) checkpoint blockade might enhance the antitumor activity of adoptively transferred CTLs. Patients and Methods Autologous MART1-specific CTLs were generated by priming with peptide-pulsed dendritic cells in the presence of interleukin-21 and enriched by peptide-major histocompatibility complex multimer-guided cell sorting. This expeditiously yielded polyclonal CTL lines uniformly expressing markers associated with an enhanced survival potential. In this first-in-human strategy, 10 patients with stage IV melanoma received the MART1-specific CTLs followed by a standard course of anti-CTLA-4 (ipilimumab). Results The toxicity profile of the combined treatment was comparable to that of ipilimumab monotherapy. Evaluation of best responses at 12 weeks yielded two continuous complete remissions, one partial response (PR) using RECIST criteria (two PRs using immune-related response criteria), and three instances of stable disease. Infused CTLs persisted with frequencies up to 2.9% of CD8+ T cells for as long as the patients were monitored (up to 40 weeks). In patients who experienced complete remissions, PRs, or stable disease, the persisting CTLs acquired phenotypic and functional characteristics of long-lived memory cells. Moreover, these patients also developed responses to nontargeted tumor antigens (epitope spreading). Conclusion We demonstrate that combining antigen-specific CTLs with CTLA-4 blockade is safe and produces durable clinical responses, likely reflecting both enhanced activity of transferred cells and improved recruitment of new responses, highlighting the promise of this strategy.


Retrovirology | 2010

In situ detection of Gag-specific CD8 + cells in the GI tract of SIV infected Rhesus macaques

Annelie Tjernlund; Jia Zhu; Kerry J. Laing; Kurt Diem; David McDonald; Julio A. Vázquez; Jianhong Cao; Claes Ohlen; M. Juliana McElrath; Louis J. Picker; Lawrence Corey

BackgroundSIV and HIV predominantly replicate in lymphoid tissue, but the study of virus specific CD8+ T cells in intact lymphoid tissue is difficult, as traditional in situ tetramer staining requires fresh tissue.ResultsIn this report, we demonstrate a novel technique using Qdot 655-conjugated peptide-MHC multimers to directly visualize SIV specific cells in cryopreserved tissue biopsies from chronically SIVmac239 infected Rhesus macaques. Qdot 655 multimers showed similar sensitivity and specificity to APC-conjugated tetramers by flow cytometry analysis, but yielded ten-fold higher signal intensity when imaged by fluorescence microscopy. Using this technique, we detected CD8+ T cells which recognize an immunodominant epitope (Gag CM9) in the spleen, lymph nodes, ileum and colon. In all these tissues, the Gag CM9 positive cells were mainly located in the extra follicular T cell zone. In the ileum and colon, we found Gag CM9 positive cells concentrated in Peyers patches and solitary lymphoid follicles, a pattern of localization not previously described.ConclusionsThe use of Qdot multimers provide an anatomic and quantitative evaluation of SIV specific CD8+ T cell responses in SIV pathogenesis, and may prove useful to studies of SIV specific CD8+ T cell responses elicited by vaccines and other immunotherapies in the non-human primate model.

Collaboration


Dive into the Jianhong Cao's collaboration.

Top Co-Authors

Avatar

M. Juliana McElrath

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Adam P. Geballe

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

John McNevin

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Cassian Yee

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

John A. Thompson

Seattle Cancer Care Alliance

View shared research outputs
Top Co-Authors

Avatar

Lawrence Corey

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Andadam P. Geballe

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Aude G. Chapuis

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Felecia Wagener

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Heather L. Sloan

Fred Hutchinson Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge