Network


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

Hotspot


Dive into the research topics where Rong Fang is active.

Publication


Featured researches published by Rong Fang.


The Journal of Infectious Diseases | 2002

An Experimental Model of Human Body Louse Infection with Rickettsia prowazekii

Linda Houhamdi; Pierre Edouard Fournier; Rong Fang; Hubert Lepidi; Didier Raoult

Rickettsia prowazekii is transmitted to humans by the body louse. A new experimental model of body louse infection with R. prowazekii is reported here. Eight hundred human lice were infected by feeding on a rabbit that was made bacteremic by injecting 2x106 plaque-forming units of R. prowazekii. The bacterium invaded the stomach cells and was released in feces, in which it was detected 5 days after infection. At day 6 after infection, as a result of the cell burst and the spread of erythrocytes in the hemolymph, the louse became bright red and died within 4 h. The life span of infected lice was shortened by 20-23 days, compared with that of uninfected control lice. Infected lice did not transmit R. prowazekii to their progeny. Through cell culture, rickettsiae were cultivated from fecal samples up to 10 days after their emission. The administration of doxycycline to the rabbit during louse feeding did not cure lice from R. prowazekii infection.


Infection and Immunity | 2007

Differential Interaction of Dendritic Cells with Rickettsia conorii: Impact on Host Susceptibility to Murine Spotted Fever Rickettsiosis

Rong Fang; Nahed Ismail; Lynn Soong; Vsevolod L. Popov; Ted Whitworth; Donald H. Bouyer; David H. Walker

ABSTRACT Spotted fever group rickettsioses are emerging and reemerging infectious diseases, some of which are life-threatening. In order to understand how dendritic cells (DCs) contribute to the host resistance or susceptibility to rickettsial diseases, we first characterized the in vitro interaction of rickettsiae with bone marrow-derived DCs (BMDCs) from resistant C57BL/6 (B6) and susceptible C3H/HeN (C3H) mice. In contrast to the exclusively cytosolic localization within endothelial cells, rickettsiae efficiently entered and localized in both phagosomes and cytosol of BMDCs from both mouse strains. Rickettsia conorii-infected BMDCs from resistant mice harbored higher bacterial loads compared to C3H mice. R. conorii infection induced maturation of BMDCs from both mouse strains as judged by upregulated expression of classical major histocompatibility complex (MHC) and costimulatory molecules. Compared to C3H counterparts, B6 BMDCs exhibited higher expression levels of MHC class II and higher interleukin-12 (IL-12) p40 production upon rickettsial infection and were more potent in priming naïve CD4+ T cells to produce gamma interferon. In vitro DC infection and T-cell priming studies suggested a delayed CD4+ T-cell activation and suppressed Th1/Th2 cell development in C3H mice. The suppressive CD4+ T-cell responses seen in C3H mice were associated with a high frequency of Foxp3+ T regulatory cells promoted by syngeneic R. conorii-infected BMDCs in the presence of IL-2. These data suggest that rickettsiae can target DCs to stimulate a protective type 1 response in resistant hosts but suppressive adaptive immunity in susceptible hosts.


Journal of Immunology | 2011

The Interaction between IL-18 and IL-18 Receptor Limits the Magnitude of Protective Immunity and Enhances Pathogenic Responses following Infection with Intracellular Bacteria

Purnima Ghose; Asim Q. Ali; Rong Fang; Digna Forbes; Billy R. Ballard; Nahed Ismail

The binding of IL-18 to IL-18Rα induces both proinflammatory and protective functions during infection, depending on the context in which it occurs. IL-18 is highly expressed in the liver of wild-type (WT) C57BL/6 mice following lethal infection with highly virulent Ixodes ovatus ehrlichia (IOE), an obligate intracellular bacterium that causes acute fatal toxic shock-like syndrome. In this study, we found that IOE infection of IL-18Rα−/− mice resulted in significantly less host cell apoptosis, decreased hepatic leukocyte recruitment, enhanced bacterial clearance, and prolonged survival compared with infected WT mice, suggesting a pathogenic role for IL-18/IL-18Rα in Ehrlichia-induced toxic shock. Although lack of IL-18R decreased the magnitude of IFN-γ producing type-1 immune response, enhanced resistance of IL-18Rα−/− mice against Ehrlichia correlated with increased proinflammatory cytokines at sites of infection, decreased systemic IL-10 production, increased frequency of protective NKT cells producing TNF-α and IFN-γ, and decreased frequency of pathogenic TNF-α–producing CD8+ T cells. Adoptive transfer of immune WT CD8+ T cells increased bacterial burden in IL-18Rα−/− mice following IOE infection. Furthermore, rIL-18 treatment of WT mice infected with mildly virulent Ehrlichia muris impaired bacterial clearance and enhanced liver injury. Finally, lack of IL-18R signal reduced dendritic cell maturation and their TNF-α production, suggesting that IL-18 might promote the adaptive pathogenic immune responses against Ehrlichia by influencing T cell priming functions of dendritic cells. Together, these results suggested that the presence or absence of IL-18R signals governs the pathogenic versus protective immunity in a model of Ehrlichia-induced immunopathology.


Clinical and Vaccine Immunology | 2003

Antigenic Classification of Rickettsia felis by Using Monoclonal and Polyclonal Antibodies

Rong Fang; Didier Raoult

ABSTRACT Rickettsia felis is a flea-transmitted rickettsia. There is a discrepancy between its reported phylogenic and phenotypic identifications. Following the first report of R. felis, it was considered by tests with serologic reagents to be closely related to another recognized flea-transmitted rickettia, R. typhi. Subsequently, it appeared to be more closely related to spotted fever group (SFG) rickettsiae by genetic analysis. In the present work, R. felis was studied by microimmunofluorescence (MIF) serologic typing and with monoclonal antibodies (MAbs). Mouse polyclonal antisera to R. felis cross-reacted only with SFG rickettsiae. A neighbor-joining analysis based on MIF indicated that R. felis is actually related to SFG rickettsiae antigenically, clustering with R. australis, R. akari, and R. montanensis. A panel of 21 MAbs was raised against a 120-kDa protein antigen or a 17-kDa polypeptide of R. felis. They cross-reacted with most members of the SFG rickettsiae but not with R. prowazekii, R. typhi, or R. canadensis of the typhus group (TG) rickettsiae. Sixty-four MAbs previously generated to seven other ricketttsial species were tested with R. felis. Three MAbs reacted with the 120-kDa antigen and were generated by R. africae, R. conorii, and R. akari, respectively. They exhibited cross-reactivities with R. felis. All our data show that R. felis harbors the antigenic profile of an SFG rickettsia.


Annals of the New York Academy of Sciences | 2003

An Experimental Model of Human Body Louse Infection with Rickettsia typhi

Linda Houhamdi; Pierre Edouard Fournier; Rong Fang; Didier Raoult

Abstract: Murine (endemic) typhus caused by Rickettsia typhi, one of the most widely distributed arthropod‐borne diseases, is transmitted to humans mainly by the oriental rat flea. The human body louse, Pediculus humanus corporis, has been suspected to have a role in the transmission of R. typhi to humans. To evaluate the potential role of Pediculus humanus corporis as a vector of murine typhus, we used R. typhi in an experimental model of human body louse infection previously used with R. prowazekii. A rabbit was made bacteremic by inoculating it with 2 × 106 plaque‐forming units of R. typhi; it remained bacteremic for at least 59 hours. Two hundred body lice infected by feeding on the bacteremic rabbit were compared to 200 uninfected control lice. Each louse population was fed once a day on the abdomen of a seronegative rabbit. On day 8 post‐infection, as a result of disruption of the gut cells and leakage of the blood meal into the hemolymph, four infected lice became bright red and died within four hours. The life span of infected lice was 20 days less than that of the controls. Infected lice did not transmit R. typhi to their progeny (eggs and larvae) as demonstrated by PCR amplification and cell culture. With an immunofluorescence assay, R. typhi was detected in feces from day 7 post‐infection, and the organism remained viable in feces for up to 80 days as demonstrated by cell culture. From the 21st day post‐infection, the rabbit used to feed the R. typhi‐infected lice developed an immunoglobulin response with a titer of 1:50 increasing to 1:200 at the 42nd day post‐infection. It showed no clinical signs of infection. The rabbit that was used to feed uninfected lice remained seronegative. Although body lice are not clearly identified vectors of R. typhi, it seems that under certain circumstances they could transmit R. typhi.


PLOS ONE | 2014

Caspase-1/ASC Inflammasome-Mediated Activation of IL-1β–ROS–NF-κB Pathway for Control of Trypanosoma cruzi Replication and Survival Is Dispensable in NLRP3−/− Macrophages

Nilay Dey; Mala Sinha; Shivali Gupta; Mariela N. González; Rong Fang; Janice J. Endsley; Bruce A. Luxon; Nisha Jain Garg

In this study, we have utilized wild-type (WT), ASC−/−, and NLRP3−/− macrophages and inhibition approaches to investigate the mechanisms of inflammasome activation and their role in Trypanosoma cruzi infection. We also probed human macrophages and analyzed published microarray datasets from human fibroblasts, and endothelial and smooth muscle cells for T. cruzi-induced changes in the expression genes included in the RT Profiler Human Inflammasome arrays. T. cruzi infection elicited a subdued and delayed activation of inflammasome-related gene expression and IL-1β production in mφs in comparison to LPS-treated controls. When WT and ASC−/− macrophages were treated with inhibitors of caspase-1, IL-1β, or NADPH oxidase, we found that IL-1β production by caspase-1/ASC inflammasome required reactive oxygen species (ROS) as a secondary signal. Moreover, IL-1β regulated NF-κB signaling of inflammatory cytokine gene expression and, subsequently, intracellular parasite replication in macrophages. NLRP3−/− macrophages, despite an inability to elicit IL-1β activation and inflammatory cytokine gene expression, exhibited a 4-fold decline in intracellular parasites in comparison to that noted in matched WT controls. NLRP3−/− macrophages were not refractory to T. cruzi, and instead exhibited a very high basal level of ROS (>100-fold higher than WT controls) that was maintained after infection in an IL-1β-independent manner and contributed to efficient parasite killing. We conclude that caspase-1/ASC inflammasomes play a significant role in the activation of IL-1β/ROS and NF-κB signaling of cytokine gene expression for T. cruzi control in human and mouse macrophages. However, NLRP3-mediated IL-1β/NFκB activation is dispensable and compensated for by ROS-mediated control of T. cruzi replication and survival in macrophages.


Infection and Immunity | 2009

CD4+ CD25+ Foxp3- T-regulatory cells produce both gamma interferon and interleukin-10 during acute severe murine spotted fever rickettsiosis.

Rong Fang; Nahed Ismail; Thomas R. Shelite; David H. Walker

ABSTRACT Spotted fever group rickettsiae cause life-threatening human infections worldwide. Until now, the immune regulatory mechanisms involved in fatal rickettsial infection have been unknown. C3H/HeN mice infected with 3 × 105 PFU of Rickettsia conorii developed an acute progressive disease, and all mice succumbed to this infection. A sublethal infection induced protective immunity, and mice survived. Compared to splenic T cells from sublethally infected mice, splenic T cells from lethally infected mice produced significantly lower levels of interleukin-2 (IL-2) and gamma interferon (IFN-γ) and a higher level of IL-10, but not of IL-4 or transforming growth factor β, and there was markedly suppressed CD4+ T-cell proliferation in response to antigen-specific stimulation with R. conorii. Furthermore, lethal infection induced significant expansion of CD4+ CD25+ Foxp3− T cells in infected organs compared to the levels in naïve and sublethally infected mice. In a lethal infection, splenic CD4+ CD25+ Foxp3− T cells, which were CTLA-4high T-bet+ and secreted both IFN-γ and IL-10, suppressed the proliferation of and IL-2 production by splenic CD4+ CD25− Foxp3− T cells in vitro. Interestingly, depletion of CD25+ T cells in vivo did not change the disease progression, but it increased the bacterial load in the lung and liver, significantly reduced the number of IFN-γ-producing Th1 cells in the spleen, and increased the serum levels of IFN-γ. These results suggested that CD4+ CD25+ T cells generated in acute murine spotted fever rickettsiosis are Th1-cell-related adaptive T-regulatory cells, which substantially contribute to suppressing the systemic immune response, possibly by a mechanism involving IL-10 and/or cytotoxic T-lymphocyte antigen 4.


PLOS ONE | 2012

Systemic treatment with cpg-b after sublethal rickettsial infection induces mouse death through indoleamine 2,3-dioxygenase (ido)

Lijun Xin; Thomas R. Shelite; Bin Gong; Nicole L. Mendell; Lynn Soong; Rong Fang; David H. Walker

Due to its strong immune stimulatory effects through TLR9, CpG-containing oligodeoxynucleotides (CpG ODN) have been tested in multiple clinical trials as vaccine adjuvant for infectious diseases and cancer. However, immune suppression induced by systemic administration of CpGs has been reported recently. In this study, we evaluated the impact of CpGs in an acute rickettsiosis model. We found that systemic treatment with type B CpG (CpG-B), but not type A CpG (CpG-A), at 2 days after sublethal R. australis infection induced mouse death. Although wild-type (WT) B6 and IDO−/− mice showed similar survival rates with three different doses of R. australis infection, treatment with CpG-B after sublethal infection consistently induced higher mortality with greater tissue bacterial loads in WT but not IDO−/− mice. Also, CpG-B treatment promoted the development of higher serum concentrations of proinflammatory cytokines/chemokines through IDO. Furthermore, while T cell-mediated immune responses enhanced by CpG-B were independent of IDO, treatment with CpG-B promoted T cell activation, PD-1 expression and cell apoptosis partially through IDO. A depletion study using anti-mPDCA-1 mAb indicated that plasmacytoid dendritic cells (pDC) were not required for CpG-B-induced death of R. australis-infected mice. Additionally, the results in iNOS−/− mice suggested that nitric oxide (NO) was partially involved in CpG-B-induced death of R. australis-infected mice. Surprisingly, pre-treatment with CpG-B before administration of a lethal dose of R. australis provided effective immunity in WT, IDO−/− and iNOS−/− mice. Taken together, our study provides evidence that CpGs exert complex immunological effects by both IDO-dependent and -independent mechanisms, and that systemic treatment with CpGs before or after infection has a significant and distinct impact on disease outcomes.


American Journal of Pathology | 2012

Contribution of NK Cells to the Innate Phase of Host Protection Against an Intracellular Bacterium Targeting Systemic Endothelium

Rong Fang; Nahed Ismail; David H. Walker

We investigated the mechanisms by which natural killer (NK) cells mediate innate host defense against infection with an endothelium-targeting intracellular bacterium, Rickettsia. We found that a robust Rickettsia-induced innate response in resistant mice cleared the bacteria early in the infection and was associated with significantly higher frequencies of splenic interferon (IFN)-γ (+) CD8(+) T cells and cytotoxic NK cells compared with susceptible mice. More importantly, NK cell-deficient Rag(-/-)γc(-/-) animals displayed significantly increased susceptibility to Rickettsia infection compared with NK cell-sufficient Rag(-/-) mice, as evidenced by impaired bacterial clearance, early development of severe thrombosis in the liver, and a decreased serum level of IFN-γ. Furthermore, the lack of NK cells also impaired host resistance of CB-17 scid mice to Rickettsia, similar to what was observed in Rag(-/-)γc(-/-) mice. Interestingly, perforin deficiency in Rag(-/-)Prf1(-/-) mice resulted in greater thrombosis and insignificantly different systemic levels of IFN-γ compared with Rag(-/-) mice, suggesting that perforin, which is mainly produced by NK cells, is involved in the prevention of vascular damage. Together, these findings reveal that NK cells mediate the innate phase of host protection against infection with rickettsiae, most likely via IFN-γ production. Furthermore, NK cells are involved in preventing rickettsial infection-induced endothelial cell damage, possibly via perforin production.


Journal of Clinical Microbiology | 2002

Detection of Rickettsia prowazekii in Body Lice and Their Feces by Using Monoclonal Antibodies

Rong Fang; Linda Houhamdi; Didier Raoult

ABSTRACT In order to identify Rickettsia prowazekii in lice, we developed a panel of 29 representative monoclonal antibodies selected from 187 positive hybridomas made by fusing splenocytes of immunized mice with SP2/0-Ag14 myeloma cells. Immunoblotting revealed that 15 monoclonal antibodies reacted with the lipopolysaccharide-like (LPS-L) antigen and 14 reacted with the epitopes of a 120-kDa protein. Only typhus group rickettsiae reacted with the monoclonal antibodies against LPS-L. R. felis, a recently identified rickettsial species, did not react with these monoclonal antibodies, confirming that it is not antigenically related to the typhus group. Monoclonal antibodies against the 120-kDa protein were highly specific for R. prowazekii. We successfully applied a selected monoclonal antibody against the 120-kDa protein to detect by immunofluorescence assay R. prowazekii in smears from 56 wild and laboratory lice, as well as in 10 samples of louse feces infected or not infected with the organism. We have developed a simple, practical, and specific diagnostic assay for clinical specimens and large-scale epidemiological surveys with a sensitivity of 91%. These monoclonal antibodies could be added to the rickettsial diagnostic panel and be used to differentiate R. prowazekii from other rickettsial species.

Collaboration


Dive into the Rong Fang's collaboration.

Top Co-Authors

Avatar

David H. Walker

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Nahed Ismail

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Claire Smalley

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Jeremy Bechelli

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Didier Raoult

Aix-Marseille University

View shared research outputs
Top Co-Authors

Avatar

Linda Houhamdi

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Bin Gong

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Tais B. Saito

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Thomas R. Shelite

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Xuemei Zhao

University of Texas Medical Branch

View shared research outputs
Researchain Logo
Decentralizing Knowledge