Network


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

Hotspot


Dive into the research topics where Rosie Jiang is active.

Publication


Featured researches published by Rosie Jiang.


Cancer Cell | 2013

A bis-Benzylidine Piperidone Targeting Proteasome Ubiquitin Receptor RPN13/ADRM1 as a therapy for cancer

Ravi K. Anchoori; Balasubramanyam Karanam; Shiwen Peng; Joshua W. Wang; Rosie Jiang; Toshihiko Tanno; Robert Z. Orlowski; William Matsui; Ming Zhao; Michelle A. Rudek; Chien Fu Hung; Xiang Chen; Kylie J. Walters; Richard Roden

The bis-benzylidine piperidone RA190 covalently binds to cysteine 88 of ubiquitin receptor RPN13 in the 19S regulatory particle and inhibits proteasome function, triggering rapid accumulation of polyubiquitinated proteins. Multiple myeloma (MM) lines, even those resistant to bortezomib, were sensitive to RA190 via endoplasmic reticulum stress-related apoptosis. RA190 stabilized targets of human papillomavirus (HPV) E6 oncoprotein, and preferentially killed HPV-transformed cells. After oral or intraperitoneal dosing of mice, RA190 distributed to plasma and major organs except the brain and inhibited proteasome function in skin and muscle. RA190 administration profoundly reduced growth of MM and ovarian cancer xenografts, and oral RA190 treatment retarded HPV16(+) syngeneic mouse tumor growth, without affecting spontaneous HPV-specific CD8(+) T cell responses, suggesting its therapeutic potential.


PLOS ONE | 2014

Impact of Inhibitors and L2 Antibodies upon the Infectivity of Diverse Alpha and Beta Human Papillomavirus Types

Kihyuck Kwak; Rosie Jiang; Joshua W. Wang; Subhashini Jagu; Reinhard Kirnbauer; Richard Roden

The licensed human papillomavirus (HPV) vaccines elicit type-restricted immunity but do not target cutaneous HPV types of the beta genus that are associated with non-melanoma skin cancer in immune-compromised patients, and it is unclear if these diverse types share a common mechanism of infection. Residues 11-88 of minor capsid protein L2 contain cross-protective epitopes, and vaccination with concatamers of this region derived from as many as eight alpha HPV (L2 α11-88x8) is being developed as an alternative prophylactic vaccine with potentially broader efficacy. There is also interest in developing broadly protective topical microbicides, such as carrageenan or heparin that block HPV receptor interactions, or small molecule inhibitors of infection. Here we have examined several inhibitors of HPV infection and antisera to L2 α11-88x8 for their breadth of activity against infection by 34 HPV types from within both the alpha and beta families using pseudovirions (PsV) carrying a luciferase reporter as surrogates for native virus. We observed that both heparin and carrageenan prevented infection by mucosatropic HPV types, but surprisingly PsV of several epidermotropic alpha4 and beta HPV types exhibited increased infectivity especially at low inhibitor concentrations. Furin and γ-secretase inhibitors and L2 α11-88x8 antiserum blocked infection by all HPV PsV types tested. These findings suggest that the distinct tropism of mucosal and cutaneous HPV may reflect distinct cell surface receptor interactions, but a common uptake mechanism dependent upon furin and γ-secretase proteolytic activities. Carrageenan, which is being tested as a vaginal microbicide, broadly inhibited infection by the high-risk mucosatropic HPV PsV, but not most skin tropic alpha and beta HPV. Vaccination with an L2 multimer derived exclusively from alpha papillomavirus sequences induced antibodies that broadly neutralized PsV of all 34 HPVs from within both the alpha and beta families, suggesting each displays conserved L2 neutralizing epitopes.


PLOS Pathogens | 2015

Immunologic Control of Mus musculus Papillomavirus Type 1

Joshua W. Wang; Rosie Jiang; Shiwen Peng; Yung Nien Chang; Chien Fu Hung; Richard Roden

Persistent papillomas developed in ~10% of out-bred immune-competent SKH-1 mice following MusPV1 challenge of their tail, and in a similar fraction the papillomas were transient, suggesting potential as a model. However, papillomas only occurred in BALB/c or C57BL/6 mice depleted of T cells with anti-CD3 antibody, and they completely regressed within 8 weeks after depletion was stopped. Neither CD4+ nor CD8+ T cell depletion alone in BALB/c or C57BL/6 mice was sufficient to permit visible papilloma formation. However, low levels of MusPV1 were sporadically detected by either genomic DNA-specific PCR analysis of local skin swabs or in situ hybridization of the challenge site with an E6/E7 probe. After switching to CD3+ T cell depletion, papillomas appeared upon 14/15 of mice that had been CD4+ T cell depleted throughout the challenge phase, 1/15 of CD8+ T cell depleted mice, and none in mice without any prior T cell depletion. Both control animals and those depleted with CD8-specific antibody generated MusPV1 L1 capsid-specific antibodies, but not those depleted with CD4-specific antibody prior to T cell depletion with CD3 antibody. Thus, normal BALB/c or C57BL/6 mice eliminate the challenge dose, whereas infection is suppressed but not completely cleared if their CD4 or CD8 T cells are depleted, and recrudescence of MusPV1 is much greater in the former following treatment with CD3 antibody, possibly reflecting their failure to generate capsid antibody. Systemic vaccination of C57BL/6 mice with DNA vectors expressing MusPV1 E6 or E7 fused to calreticulin elicits potent CD8 T cell responses and these immunodominant CD8 T cell epitopes were mapped. Adoptive transfer of a MusPV1 E6-specific CD8+ T cell line controlled established MusPV1 infection and papilloma in RAG1-knockout mice. These findings suggest the potential of immunotherapy for HPV-related disease and the importance of host immunogenetics in the outcome of infection.


PLOS ONE | 2013

Multivalent Human Papillomavirus L1 DNA Vaccination Utilizing Electroporation

Kihyuck Kwak; Rosie Jiang; Subhashini Jagu; Joshua W. Wang; Chenguang Wang; Neil D. Christensen; Richard Roden

Objectives Naked DNA vaccines can be manufactured simply and are stable at ambient temperature, but require improved delivery technologies to boost immunogenicity. Here we explore in vivo electroporation for multivalent codon-optimized human papillomavirus (HPV) L1 and L2 DNA vaccination. Methods Balb/c mice were vaccinated three times at two week intervals with a fusion protein comprising L2 residues ∼11−88 of 8 different HPV types (11−88×8) or its DNA expression vector, DNA constructs expressing L1 only or L1+L2 of a single HPV type, or as a mixture of several high-risk HPV types and administered utilizing electroporation, i.m. injection or gene gun. Serum was collected two weeks and 3 months after the last vaccination. Sera from immunized mice were tested for in-vitro neutralization titer, and protective efficacy upon passive transfer to naive mice and vaginal HPV challenge. Heterotypic interactions between L1 proteins of HPV6, HPV16 and HPV18 in 293TT cells were tested by co-precipitation using type-specific monoclonal antibodies. Results Electroporation with L2 multimer DNA did not elicit detectable antibody titer, whereas DNA expressing L1 or L1+L2 induced L1-specific, type-restricted neutralizing antibodies, with titers approaching those induced by Gardasil. Co-expression of L2 neither augmented L1-specific responses nor induced L2-specific antibodies. Delivery of HPV L1 DNA via in vivo electroporation produces a stronger antibody response compared to i.m. injection or i.d. ballistic delivery via gene gun. Reduced neutralizing antibody titers were observed for certain types when vaccinating with a mixture of L1 (or L1+L2) vectors of multiple HPV types, likely resulting from heterotypic L1 interactions observed in co-immunoprecipitation studies. High titers were restored by vaccinating with individual constructs at different sites, or partially recovered by co-expression of L2, such that durable protective antibody titers were achieved for each type. Discussion Multivalent vaccination via in vivo electroporation requires spatial separation of individual type L1 DNA vaccines.


Expert Review of Vaccines | 2016

Progress and prospects for L2-based human papillomavirus vaccines

Rosie Jiang; Christina Schellenbacher; Bryce Chackerian; Richard Roden

ABSTRACT Human papillomavirus (HPV) is a worldwide public health problem, particularly in resource-limited countries. Fifteen high-risk genital HPV types are sexually transmitted and cause 5% of all cancers worldwide, primarily cervical, anogenital and oropharyngeal carcinomas. Skin HPV types are generally associated with benign disease, but a subset is linked to non-melanoma skin cancer. Licensed HPV vaccines based on virus-like particles (VLPs) derived from L1 major capsid antigen of key high risk HPVs are effective at preventing these infections but do not cover cutaneous types and are not therapeutic. Vaccines targeting L2 minor capsid antigen, some using capsid display, adjuvant and fusions with early HPV antigens or Toll-like receptor agonists, are in development to fill these gaps. Progress and challenges with L2-based vaccines are summarized.


Journal of Virology | 2017

Spontaneous and Vaccine-Induced Clearance of Mus Musculus Papillomavirus 1 Infection

Rosie Jiang; Joshua W. Wang; Shiwen Peng; Tsui Chin Huang; Chenguang Wang; Fabiana Cannella; Yung Nien Chang; Raphael P. Viscidi; Simon R. Best; Chien Fu Hung; Richard Roden

ABSTRACT Mus musculus papillomavirus 1 (MmuPV1/MusPV1) induces persistent papillomas in immunodeficient mice but not in common laboratory strains. To facilitate the study of immune control, we sought an outbred and immunocompetent laboratory mouse strain in which persistent papillomas could be established. We found that challenge of SKH1 mice (Crl:SKH1-Hrhr) with MmuPV1 by scarification on their tail resulted in three clinical outcomes: (i) persistent (>2-month) papillomas (∼20%); (ii) transient papillomas that spontaneously regress, typically within 2 months (∼15%); and (iii) no visible papillomas and viral clearance (∼65%). SKH1 mice with persistent papillomas were treated by using a candidate preventive/therapeutic naked-DNA vaccine that expresses human calreticulin (hCRT) fused in frame to MmuPV1 E6 (mE6) and mE7 early proteins and residues 11 to 200 of the late protein L2 (hCRTmE6/mE7/mL2). Three intramuscular DNA vaccinations were delivered biweekly via in vivo electroporation, and both humoral and CD8 T cell responses were mapped and measured. Previously persistent papillomas disappeared within 2 months after the final vaccination. Coincident virologic clearance was confirmed by in situ hybridization and a failure of disease to recur after CD3 T cell depletion. Vaccination induced strong mE6 and mE7 CD8+ T cell responses in all mice, although they were significantly weaker in mice that initially presented with persistent warts than in those that spontaneously cleared their infection. A human papillomavirus 16 (HPV16)-targeted version of the DNA vaccine also induced L2 antibodies and protected mice from vaginal challenge with an HPV16 pseudovirus. Thus, MmuPV1 challenge of SKH1 mice is a promising model of spontaneous and immunotherapy-directed clearances of HPV-related disease. IMPORTANCE High-risk-type human papillomaviruses (hrHPVs) cause 5% of all cancer cases worldwide, notably cervical, anogenital, and oropharyngeal cancers. Since preventative HPV vaccines have not been widely used in many countries and do not impact existing infections, there is considerable interest in the development of therapeutic vaccines to address existing disease and infections. The strict tropism of HPV requires the use of animal papillomavirus models for therapeutic vaccine development. However, MmuPV1 failed to grow in common laboratory strains of mice with an intact immune system. We show that MmuPV1 challenge of the outbred immunocompetent SKH1 strain produces both transient and persistent papillomas and that vaccination of the mice with a DNA expressing an MmuPV1 E6E7L2 fusion with calreticulin can rapidly clear persistent papillomas. Furthermore, an HPV16-targeted version of the DNA can protect against vaginal challenge with HPV16, suggesting the promise of this approach to both prevent and treat papillomavirus-related disease.


Journal of Ovarian Research | 2017

Early and consistent overexpression of ADRM1 in ovarian high-grade serous carcinoma

Rosie Jiang; Anna Yemelyanova; Deyin Xing; Ravi K. Anchoori; Jun Hamazaki; Shigeo Murata; Jeffrey D. Seidman; Tian Li Wang; Richard Roden

BackgroundOvarian carcinoma is highly dependent on the ubiquitin proteasome system (UPS), but its clinical response to treatment with the proteasome inhibitor bortezomib has been disappointing. This has driven exploration of alternate approaches to target the UPS in ovarian cancer. Recently, proteasome inhibitors targeting the 19S regulatory particle-associated RPN13 protein have been described, such as RA190. RPN13, which is encoded by ADRM1, facilitates the recognition by the proteasome of its polyubiquinated substrates. Inhibition of RPN13 produces a rapid, toxic accumulation of polyubiquitinated proteins in ovarian and other cancer cells, triggering apoptosis.Here, we sought to determine if RPN13 is available as a target in precursors of ovarian/fallopian tube cancer as well as all advanced cases, and the impact of increased ADRM1 gene copy number on sensitivity of ovarian cancer to RA190.MethodsADRM1 mRNA was quantified by RNAscope in situ hybridization and RPN13 protein detected by immunohistochemistry in high grade serous carcinoma (HGSC) of the ovary and serous tubal intraepithelial carcinoma (STIC). Amplification of ADRM1 and sensitivity to RA190 were determined in ovarian cancer cell lines.ResultsHere, we demonstrate that expression of ADRM1mRNA is significantly elevated in STIC and HGSC as compared to normal fallopian tube epithelium. ADRM1 mRNA and RPN13 were ubiquitously and robustly expressed in ovarian carcinoma tissue and cell lines. No correlation was found between ADRM1 amplification and sensitivity of ovarian cancer cell lines to RA190, but all were susceptible.ConclusionsRPN13 can potentially be targeted by RA190 in both in situ and metastatic ovarian carcinoma. Ovarian cancer cell lines are sensitive to RA190 regardless of whether the ADRM1 gene is amplified.


Cancer immunology research | 2018

Integration of Oncogenes via Sleeping Beauty as a Mouse Model of HPV16+ Oral Tumors and Immunologic Control

Yi Hsin Lin; Ming Chieh Yang; Ssu Hsueh Tseng; Rosie Jiang; Andrew Yang; Emily Farmer; Shiwen Peng; Talia Henkle; Yung Nien Chang; Chien Fu Hung; T. C. Wu

A preclinical mouse model of spontaneous, HPV+ oral tumors was generated. Sleeping beauty transposase system-mediated oncogene transfection was assisted by electroporation. This model allowed for the evaluation of disease intervention efficacy and analysis of tumor cell migration. Human papillomavirus type 16 (HPV16) is the etiologic factor for cervical cancer and a subset of oropharyngeal cancers. Although several prophylactic HPV vaccines are available, no effective therapeutic strategies to control active HPV diseases exist. Tumor implantation models are traditionally used to study HPV-associated buccal tumors. However, they fail to address precancerous phases of disease progression and display tumor microenvironments distinct from those observed in patients. Previously, K14-E6/E7 transgenic mouse models have been used to generate spontaneous tumors. However, the rate of tumor formation is inconsistent, and the host often develops immune tolerance to the viral oncoproteins. We developed a preclinical, spontaneous, HPV16+ buccal tumor model using submucosal injection of oncogenic plasmids expressing HPV16-E6/E7, NRasG12V, luciferase, and sleeping beauty (SB) transposase, followed by electroporation in the buccal mucosa. We evaluated responses to immunization with a pNGVL4a-CRT/E7(detox) therapeutic HPV DNA vaccine and tumor cell migration to distant locations. Mice transfected with plasmids encoding HPV16-E6/E7, NRasG12V, luciferase, and SB transposase developed tumors within 3 weeks. We also found transient anti-CD3 administration is required to generate tumors in immunocompetent mice. Bioluminescence signals from luciferase correlated strongly with tumor growth, and tumors expressed HPV16-associated markers. We showed that pNGVL4a-CRT/E7(detox) administration resulted in antitumor immunity in tumor-bearing mice. Lastly, we demonstrated that the generated tumor could migrate to tumor-draining lymph nodes. Our model provides an efficient method to induce spontaneous HPV+ tumor formation, which can be used to identify effective therapeutic interventions, analyze tumor migration, and conduct tumor biology research. Cancer Immunol Res; 6(3); 305–19. ©2018 AACR.


ACS Omega | 2018

Covalent Rpn13-Binding Inhibitors for the Treatment of Ovarian Cancer

Ravi K. Anchoori; Rosie Jiang; Shiwen Peng; Ruey-shyang Soong; Aliyah Algethami; Michelle A. Rudek; Nicole M. Anders; Chien Fu Hung; Xiang Chen; Xiuxiu Lu; Olumide Kayode; Marzena Dyba; Kylie J. Walters; Richard Roden

Substitution of the m,p-chloro groups of bis-benzylidinepiperidone RA190 for p-nitro, generating RA183, enhanced covalent drug binding to Cys88 of RPN13. Treatment of cancer cell lines with RA183 inhibited ubiquitin-mediated protein degradation, resulting in rapid accumulation of high-molecular-weight polyubiquitinated proteins, blockade of NFκB signaling, endoplasmic reticulum stress, an unfolded protein response, production of reactive oxygen species, and apoptotic cell death. High-grade ovarian cancer, triple-negative breast cancer, and multiple myeloma cell lines were particularly vulnerable to RA183. RA183 stabilized a tetraubiquitin-linked firefly luciferase reporter protein in cancer cell lines and mice, demonstrating in vitro and in vivo proteasomal inhibition, respectively. However, RA183 was rapidly cleared from plasma, likely reflecting its rapid degradation to the active compound RA9, as seen in human liver microsomes. Intraperitoneal administration of RA183 inhibited proteasome function and orthotopic tumor growth in mice bearing human ovarian cancer model ES2-luc ascites or syngeneic ID8-luc tumor.


Clinical Cancer Research | 2016

Abstract A74: Proteasome subunit RPN13 as a candidate target for treatment of ovarian carcinoma.

Rosie Jiang; Ravi K. Anchoori; Anna V. Yemelyanova; Chien Fu Hung

The aberrant metabolism of ovarian cancer cells produces an excess of misfolded proteins and particular sensitivity to proteasome inhibition. Blockade of proteasome 19S regulatory particle (RP) deubiquitinase and/or 20S subunit proteolytic function results in a rapid buildup of denatured protein aggregates and triggers a p53-independent cell death. However, treatment of ovarian cancer with the licensed proteasome inhibitor bortezomib did not produce sufficient benefit to warrant further development. This setback led us to develop second generation inhibitors representing a distinct class of molecule and new mechanism of proteasome inhibition. We have developed novel bis-benzylidine piperidones, RA183 and RA190, which bind covalently to the RPN13 subunit of the 19S RP. RPN13 acts as an ubiquitin receptor and promotes the deubiquitinase activity of its binding partner UCH37. RA183 and RA190 cause rapid accumulation of very high molecular weight poly-ubiquitinated proteins, higher and more rapid than seen with bortezomib treatment, suggesting they inhibit RPN13 function, and thus deubiquitination by the 19S regulatory particle and 20S catalytic subunit activity. The accumulation of polyubiquitinated proteins triggered by RA183 or RA190 causes endoplasmic reticulum (ER) stress and an unresolved unfolded protein response that rapidly triggers apoptosis. RA190 and RA183 are active against multiple high-grade ovarian carcinoma cell lines in vitro and ovarian cancer models in mice. In nude mice carrying intraperitoneal ES2-luciferase xenografts, RA190 treatment (10mg/kg) significantly inhibited tumor growth. RPN13 is amplified in a subset of high grade ovarian cancers (10-20%), but its amplification did not correlate with a change in the sensitivity of ovarian carcinoma cell lines to RPN13 inhibitor. Sensitivity to RA190 also did not correlate with the level of pre-treatment stress or protein poly-ubiquitination. Knockdown of RPN13 expression in mouse ovarian cell line, ID8, increased its sensitivity to RA190. However attempts to generate knockout of RPN13 by CRISPR in NIH:OVCAR-3 failed and appeared to reflect toxicity. RPN13 mRNA was detected by chromogenic RNA in situ hybridization in all high grade serous carcinoma (HGSC) cases tested and matched precursor serous tubal intraepithelial carcinomas(n=16), and was overexpressed in all relative to adjacent normal fallopian tube epithelium. We conclude that RPN13 has promise as a target for treatment of ovarian carcinomas and possibly HGSC precursor STIC lesions, and that further development of RPN13 inhibitors warranted. Citation Format: Rosie T. Jiang, Ravi K. Anchoori, Anna V. Yemelyanova, Chien-fu Hung. Proteasome subunit RPN13 as a candidate target for treatment of ovarian carcinoma. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Ovarian Cancer Research: Exploiting Vulnerabilities; Oct 17-20, 2015; Orlando, FL. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(2 Suppl):Abstract nr A74.

Collaboration


Dive into the Rosie Jiang's collaboration.

Top Co-Authors

Avatar

Richard Roden

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Chien Fu Hung

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Joshua W. Wang

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Shiwen Peng

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chenguang Wang

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Kihyuck Kwak

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge