Network


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

Hotspot


Dive into the research topics where Huanle Luo is active.

Publication


Featured researches published by Huanle Luo.


Nature Medicine | 2017

A live-attenuated Zika virus vaccine candidate induces sterilizing immunity in mouse models

Chao Shan; Antonio E. Muruato; Bruno T.D. Nunes; Huanle Luo; Xuping Xie; Daniele Barbosa de Almeida Medeiros; Maki Wakamiya; Robert B. Tesh; Alan D. T. Barrett; Tian Wang; Scott C. Weaver; Pedro Fernando da Costa Vasconcelos; Shannan L. Rossi; Pei Yong Shi

Zika virus (ZIKV) infection of pregnant women can cause a wide range of congenital abnormalities, including microcephaly, in the infant, a condition now collectively known as congenital ZIKV syndrome. A vaccine to prevent or significantly attenuate viremia in pregnant women who are residents of or travelers to epidemic or endemic regions is needed to avert congenital ZIKV syndrome, and might also help to suppress epidemic transmission. Here we report on a live-attenuated vaccine candidate that contains a 10-nucleotide deletion in the 3′ untranslated region of the ZIKV genome (10-del ZIKV). The 10-del ZIKV is highly attenuated, immunogenic, and protective in type 1 interferon receptor–deficient A129 mice. Crucially, a single dose of 10-del ZIKV induced sterilizing immunity with a saturated neutralizing antibody titer, which no longer increased after challenge with an epidemic ZIKV, and completely prevented viremia. The immunized mice also developed a robust T cell response. Intracranial inoculation of 1-d-old immunocompetent CD-1 mice with 1 × 104 infectious focus units (IFU) of 10-del ZIKV caused no mortality, whereas infections with 10 IFU of wild-type ZIKV were lethal. Mechanistically, the attenuated virulence of 10-del ZIKV may be due to decreased viral RNA synthesis and increased sensitivity to type-1-interferon inhibition. The attenuated 10-del ZIKV was incapable of infecting mosquitoes after oral feeding of spiked-blood meals, representing an additional safety feature. Collectively, the safety and efficacy results suggest that further development of this promising, live-attenuated ZIKV vaccine candidate is warranted.


Cell | 2017

Vaccine Mediated Protection Against Zika Virus-Induced Congenital Disease

Justin M. Richner; Brett W. Jagger; Chao Shan; Camila R. Fontes; Kimberly A. Dowd; Bin Cao; Sunny Himansu; Elizabeth A. Caine; Bruno T.D. Nunes; Daniele Barbosa de Almeida Medeiros; Antonio E. Muruato; Bryant M. Foreman; Huanle Luo; Tian Wang; Alan D. T. Barrett; Scott C. Weaver; Pedro Fernando da Costa Vasconcelos; Shannan L. Rossi; Giuseppe Ciaramella; Indira U. Mysorekar; Theodore C. Pierson; Pei Yong Shi; Michael S. Diamond

The emergence of Zika virus (ZIKV) and its association with congenital malformations has prompted the rapid development of vaccines. Although efficacy with multiple viral vaccine platforms has been established in animals, no study has addressed protection during pregnancy. We tested in mice two vaccine platforms, a lipid nanoparticle-encapsulated modified mRNA vaccine encoding ZIKV prM and E genes and a live-attenuated ZIKV strain encoding an NS1 protein without glycosylation, for their ability to protect against transmission to the fetus. Vaccinated dams challenged with a heterologous ZIKV strain at embryo day 6 (E6) and evaluated at E13 showed markedly diminished levels of viral RNA in maternal, placental, and fetal tissues, which resulted in protection against placental damage and fetal demise. As modified mRNA and live-attenuated vaccine platforms can restrict in utero transmission of ZIKV in mice, their further development in humans to prevent congenital ZIKV syndrome is warranted.


Nature Medicine | 2017

A chikungunya fever vaccine utilizing an insect-specific virus platform

Jesse H. Erasmus; Albert J. Auguste; Jason T. Kaelber; Huanle Luo; Shannan L. Rossi; Karla A. Fenton; Grace Leal; Dal Young Kim; Wah Chiu; Tian Wang; Ilya Frolov; Farooq Nasar; Scott C. Weaver

Traditionally, vaccine development involves tradeoffs between immunogenicity and safety. Live-attenuated vaccines typically offer rapid and durable immunity but have reduced safety when compared to inactivated vaccines. In contrast, the inability of inactivated vaccines to replicate enhances safety at the expense of immunogenicity, often necessitating multiple doses and boosters. To overcome these tradeoffs, we developed the insect-specific alphavirus, Eilat virus (EILV), as a vaccine platform. To address the chikungunya fever (CHIKF) pandemic, we used an EILV cDNA clone to design a chimeric virus containing the chikungunya virus (CHIKV) structural proteins. The recombinant EILV/CHIKV was structurally identical at 10 Å to wild-type CHIKV, as determined by single-particle cryo-electron microscopy, and it mimicked the early stages of CHIKV replication in vertebrate cells from attachment and entry to viral RNA delivery. Yet the recombinant virus remained completely defective for productive replication, providing a high degree of safety. A single dose of EILV/CHIKV produced in mosquito cells elicited rapid (within 4 d) and long-lasting (>290 d) neutralizing antibodies that provided complete protection in two different mouse models. In nonhuman primates, EILV/CHIKV elicited rapid and robust immunity that protected against viremia and telemetrically monitored fever. Our EILV platform represents the first structurally native application of an insect-specific virus in preclinical vaccine development and highlights the potential application of such viruses in vaccinology.


Cell Reports | 2017

Functional Analysis of Glycosylation of Zika Virus Envelope Protein

Camila R. Fontes-Garfias; Chao Shan; Huanle Luo; Antonio E. Muruato; Daniele Barbosa de Almeida Medeiros; Elizabeth Mays; Xuping Xie; Jing Zou; Christopher M. Roundy; Maki Wakamiya; Shannan L. Rossi; Tian Wang; Scott C. Weaver; Pei Yong Shi

Zika virus (ZIKV) infection causes devastating congenital abnormities and Guillain-Barré syndrome. The ZIKV envelope (E) protein is responsible for viral entry and represents a major determinant for viral pathogenesis. Like other flaviviruses, the ZIKV E protein is glycosylated at amino acid N154. To study the function of E glycosylation, we generated a recombinant N154Q ZIKV that lacks the E glycosylation and analyzed the mutant virus in mammalian and mosquito hosts. In mouse models, the mutant was attenuated, as evidenced by lower viremia, decreased weight loss, and no mortality; however, knockout of E glycosylation did not significantly affect neurovirulence. Mice immunized with the mutant virus developed a robust neutralizing antibody response and were completely protected from wild-type ZIKV challenge. In mosquitoes, the mutant virus exhibited diminished oral infectivity for the Aedes aegypti vector. Collectively, the results demonstrate that E glycosylation is critical for ZIKV infection of mammalian and mosquito hosts.


F1000Research | 2016

West Nile Virus Infection in the Central Nervous System

Evandro R. Winkelmann; Huanle Luo; Tian Wang

West Nile virus (WNV), a neurotropic single-stranded flavivirus has been the leading cause of arboviral encephalitis worldwide. Up to 50% of WNV convalescent patients in the United States were reported to have long-term neurological sequelae. Neither antiviral drugs nor vaccines are available for humans. Animal models have been used to investigate WNV pathogenesis and host immune response in humans. In this review, we will discuss recent findings from studies in animal models of WNV infection, and provide new insights on WNV pathogenesis and WNV-induced host immunity in the central nervous system.


Nature Communications | 2018

An evolutionary NS1 mutation enhances Zika virus evasion of host interferon induction

Hongjie Xia; Huanle Luo; Chao Shan; Antonio E. Muruato; Bruno T.D. Nunes; Daniele Barbosa de Almeida Medeiros; Jing Zou; Xuping Xie; Maria Isabel Giraldo; Pedro Fernando da Costa Vasconcelos; Scott C. Weaver; Tian Wang; Ricardo Rajsbaum; Pei Yong Shi

Virus–host interactions determine an infection outcome. The Asian lineage of Zika virus (ZIKV), responsible for the recent epidemics, has fixed a mutation in the NS1 gene after 2012 that enhances mosquito infection. Here we report that the same mutation confers NS1 to inhibit interferon-β induction. This mutation enables NS1 binding to TBK1 and reduces TBK1 phosphorylation. Engineering the mutation into a pre-epidemic ZIKV strain debilitates the virus for interferon-β induction; reversing the mutation in an epidemic ZIKV strain invigorates the virus for interferon-β induction; these mutational effects are lost in IRF3-knockout cells. Additionally, ZIKV NS2A, NS2B, NS4A, NS4B, and NS5 can also suppress interferon-β production through targeting distinct components of the RIG-I pathway; however, for these proteins, no antagonistic difference is observed among various ZIKV strains. Our results support the mechanism that ZIKV has accumulated mutation(s) that increases the ability to evade immune response and potentiates infection and epidemics.The Asian lineage of Zika virus (ZIKV) has acquired a mutation in NS1 that enhances mosquito infection. Here, Xia et al. show that the same mutation interferes with interferon production through interaction with TBK1 and affects ZIKV replication in mice.


Journal of Virology | 2016

Dysregulation of Toll-Like Receptor 7 Compromises Innate and Adaptive T Cell Responses and Host Resistance to an Attenuated West Nile Virus Infection in Old Mice

Guorui Xie; Huanle Luo; Lan Pang; Bi Hung Peng; Evandro R. Winkelmann; Brenna McGruder; Joseph Hesse; Melissa C. Whiteman; Gerald A. Campbell; Gregg N. Milligan; Yingzi Cong; Alan D. T. Barrett; Tian Wang

ABSTRACT The elderly are known to have enhanced susceptibility to infections and an impaired capacity to respond to vaccination. West Nile virus (WNV), a mosquito-borne flavivirus, has induced severe neurological symptoms, mostly in the elderly population. No vaccines are available for human use. Recent work showed that an attenuated WNV, a nonstructural (NS) 4B-P38G mutant, induced no lethality but strong immune responses in young (6- to 10-week-old) mice. While studying protective efficacy, we found unexpectedly that old (21- to 22-month) mice were susceptible to WNV NS4B-P38G mutant infection but were protected from subsequent lethal wild-type WNV challenge. Compared to responses in young mice, the NS4B-P38G mutant triggered higher inflammatory cytokine and interleukin-10 (IL-10) production, a delayed γδ T cell expansion, and lower antibody and WNV-specific T cell responses in old mice. Toll-like receptor 7 (TLR7) is expressed on multiple types of cells. Impaired TLR7 signaling in old mice led to dendritic cell (DC) antigen-presenting function compromise and a reduced γδ T cell and regulatory T cell (Treg) expansion during NS4B-P38G mutant infection. R848, a TLR7 agonist, decreased host vulnerability in NS4B-P38G-infected old mice by enhancing γδ T cell and Treg expansion and the antigen-presenting capacity of DCs, thereby promoting T cell responses. In summary, our results suggest that dysregulation of TLR7 partially contributes to impaired innate and adaptive T cell responses and an enhanced vulnerability in old mice during WNV NS4B-P38G mutant infection. R848 increases the safety and efficacy during immunization of old mice with the WNV NS4B-P38G mutant. IMPORTANCE The elderly are known to have enhanced susceptibility to infections and an impaired capacity to respond to vaccination. West Nile virus (WNV), an emerging mosquito-borne flavivirus, has induced severe neurological symptoms more frequently in the elderly population. No vaccines are available for human use. Here, we used an aged mouse model to investigate the protective efficacy of an attenuated WNV, the nonstructural 4B-P38G mutant, which was previously shown to induce no lethality but strong immune responses in young adult mice. Studies that contribute to a mechanistic understanding of immune defects in the elderly will allow the development of strategies to improve responses to infectious diseases and to increase vaccine efficacy and safety in aging individuals.


Journal of Immunology | 2017

TLR7 Signaling Regulates Th17 Cells and Autoimmunity: Novel Potential for Autoimmune Therapy

Jian Ye; Yadan Wang; Xia Liu; Lingyun Li; Adeleye Opejin; Eddy C. Hsueh; Huanle Luo; Tian Wang; Daniel Hawiger; Guangyong Peng

Innate regulation through TLR signaling has been shown to be important for promoting T cell subset development and function. However, limited information is known about whether differential TLR signaling can selectively inhibit Th17 and/or Th1 cells, which are important for controlling excessive inflammation and autoimmune responses. In this article, we demonstrate that activation of TLR7 signaling in T cells can inhibit Th17 cell differentiation from naive T cells and IL-17 production in established Th17 cells. We further report that downregulation of STAT3 signaling is responsible for TLR7-mediated inhibition of Th17 cells due to induction of suppressor of cytokine signaling 3 and 5. TLR7-mediated suppression of Th17 cells does not require dendritic cell involvement. In addition, we show that TLR7 signaling can suppress Th1 cell development and function through a mechanism different from Th17 cell suppression. Importantly, our complementary in vivo studies demonstrate that treatment with the TLR7 ligand imiquimod can inhibit Th1 and Th17 cells, resulting in the prevention of, and an immunotherapeutic reduction in, experimental autoimmune encephalomyelitis. These studies identify a new strategy to manipulate Th17/Th1 cells through TLR7 signaling, with important implications for successful immunotherapy against autoimmune and inflammatory diseases.


Vaccine | 2015

A West Nile virus NS4B-P38G mutant strain induces cell intrinsic innate cytokine responses in human monocytic and macrophage cells.

Guorui Xie; Huanle Luo; Bing Tian; Brian R. Mann; Xiaoyong Bao; Jere W. McBride; Robert B. Tesh; Alan D. T. Barrett; Tian Wang

Previous studies have shown that an attenuated West Nile virus (WNV) nonstructural (NS) 4B-P38G mutant induces stronger innate and adaptive immune responses than wild-type WNV in mice, which has important applications to vaccine development. To investigate the mechanism of immunogenicity, we characterized WNV NS4B-P38G mutant infection in two human cell lines-THP-1 cells and THP-1 macrophages. Although the NS4B-P38G mutant produced more viral RNA than the parental WNV NY99 in both cell types, there was no detectable infectious virus in the supernatant of either cell type. Nonetheless, the attenuated mutant boosted higher innate cytokine responses than virulent parental WNV NY99 in these cells. The NS4B-P38G mutant infection of THP-1 cells led to more diverse and robust innate cytokine responses than that seen in THP-1 macrophages, which were mediated by toll-like receptor (TLR)7 and retinoic acid-inducible gene 1(RIG-I) signaling pathways. Overall, these results suggest that a defective viral life cycle during NS4B-P38G mutant infection in human monocytic and macrophage cells leads to more potent cell intrinsic innate cytokine responses.


F1000Research | 2018

Recent advances in understanding West Nile virus host immunity and viral pathogenesis

Huanle Luo; Tian Wang

West Nile virus (WNV), a mosquito-borne flavivirus, has been a significant public health concern in the United States for nearly two decades. The virus has been linked to acute viral encephalitis, neurological sequelae, and chronic kidney diseases. Neither antiviral drugs nor vaccines are currently available for humans. In vitro cell culture and experimental animal models have been used to study WNV infection in humans. In this review, we will focus on recent findings and provide new insights into WNV host immunity and viral pathogenesis.

Collaboration


Dive into the Huanle Luo's collaboration.

Top Co-Authors

Avatar

Tian Wang

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Alan D. T. Barrett

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Scott C. Weaver

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Chao Shan

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Pei Yong Shi

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Antonio E. Muruato

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Evandro R. Winkelmann

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Guorui Xie

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Robert B. Tesh

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Shannan L. Rossi

University of Texas Medical Branch

View shared research outputs
Researchain Logo
Decentralizing Knowledge