Network


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

Hotspot


Dive into the research topics where Frederic Bibollet-Ruche is active.

Publication


Featured researches published by Frederic Bibollet-Ruche.


Cell | 2006

Nef-Mediated Suppression of T Cell Activation Was Lost in a Lentiviral Lineage that Gave Rise to HIV-1

Michael Schindler; Jan Münch; Olaf Kutsch; Hui Li; Mario L. Santiago; Frederic Bibollet-Ruche; Michaela Müller-Trutwin; Francis J. Novembre; Martine Peeters; Valérie Courgnaud; Elizabeth Bailes; Pierre Roques; Donald L. Sodora; Guido Silvestri; Paul M. Sharp; Beatrice H. Hahn; Frank Kirchhoff

High-level immune activation and T cell apoptosis represent a hallmark of HIV-1 infection that is absent from nonpathogenic SIV infections in natural primate hosts. The mechanisms causing these varying levels of immune activation are not understood. Here, we report that nef alleles from the great majority of primate lentiviruses, including HIV-2, downmodulate TCR-CD3 from infected T cells, thereby blocking their responsiveness to activation. In contrast, nef alleles from HIV-1 and a subset of closely related SIVs fail to downregulate TCR-CD3 and to inhibit cell death. Thus, Nef-mediated suppression of T cell activation is a fundamental property of primate lentiviruses that likely evolved to maintain viral persistence in the context of an intact host immune system. This function was lost during viral evolution in a lineage that gave rise to HIV-1 and may have predisposed the simian precursor of HIV-1 for greater pathogenicity in humans.


Cell Host & Microbe | 2009

Tetherin-Driven Adaptation of Vpu and Nef Function and the Evolution of Pandemic and Nonpandemic HIV-1 Strains

Daniel Sauter; Michael Schindler; Anke Specht; Wilmina N. Landford; Jan Münch; Kyeong-Ae Kim; Jörg Votteler; Ulrich S. Schubert; Frederic Bibollet-Ruche; Brandon F. Keele; Jun Takehisa; Yudelca Ogando; Christina Ochsenbauer; John C. Kappes; Ahidjo Ayouba; Martine Peeters; Gerald H. Learn; George M. Shaw; Paul M. Sharp; Paul D. Bieniasz; Beatrice H. Hahn; Theodora Hatziioannou; Frank Kirchhoff

Vpu proteins of pandemic HIV-1 M strains degrade the viral receptor CD4 and antagonize human tetherin to promote viral release and replication. We show that Vpus from SIVgsn, SIVmus, and SIVmon infecting Cercopithecus primate species also degrade CD4 and antagonize tetherin. In contrast, SIVcpz, the immediate precursor of HIV-1, whose Vpu shares a common ancestry with SIVgsn/mus/mon Vpu, uses Nef rather than Vpu to counteract chimpanzee tetherin. Human tetherin, however, is resistant to Nef and thus poses a significant barrier to zoonotic transmission of SIVcpz to humans. Remarkably, Vpus from nonpandemic HIV-1 O strains are poor tetherin antagonists, whereas those from the rare group N viruses do not degrade CD4. Thus, only HIV-1 M evolved a fully functional Vpu following the three independent cross-species transmissions that resulted in HIV-1 groups M, N, and O. This may explain why group M viruses are almost entirely responsible for the global HIV/AIDS pandemic.


Journal of Experimental Medicine | 2005

Antigenic conservation and immunogenicity of the HIV coreceptor binding site

Julie M. Decker; Frederic Bibollet-Ruche; Xiping Wei; Shuyi Wang; David N. Levy; Wenquan Wang; Eric Delaporte; Martine Peeters; Cynthia A. Derdeyn; Susan Allen; Eric Hunter; Michael S. Saag; James A. Hoxie; Beatrice H. Hahn; Peter D. Kwong; James E. Robinson; George M. Shaw

Immunogenic, broadly reactive epitopes of the HIV-1 envelope glycoprotein could serve as important targets of the adaptive humoral immune response in natural infection and, potentially, as components of an acquired immune deficiency syndrome vaccine. However, variability in exposed epitopes and a combination of highly effective envelope-cloaking strategies have made the identification of such epitopes problematic. Here, we show that the chemokine coreceptor binding site of HIV-1 from clade A, B, C, D, F, G, and H and circulating recombinant form (CRF)01, CRF02, and CRF11, elicits high titers of CD4-induced (CD4i) antibody during natural human infection and that these antibodies bind and neutralize viruses as divergent as HIV-2 in the presence of soluble CD4 (sCD4). 178 out of 189 (94%) HIV-1–infected patients had CD4i antibodies that neutralized sCD4-pretreated HIV-2 in titers (50% inhibitory concentration) as high as 1:143,000. CD4i monoclonal antibodies elicited by HIV-1 infection also neutralized HIV-2 pretreated with sCD4, and polyclonal antibodies from HIV-1–infected humans competed specifically with such monoclonal antibodies for binding. In vivo, variants of HIV-1 with spontaneously exposed coreceptor binding surfaces were detected in human plasma; these viruses were neutralized directly by CD4i antibodies. Despite remarkable evolutionary diversity among primate lentiviruses, functional constraints on receptor binding create opportunities for broad humoral immune recognition, which in turn serves to constrain the viral quasispecies.


PLOS Pathogens | 2008

Molecular ecology and natural history of Simian foamy virus infection in wild-living chimpanzees

Weimin Liu; Michael Worobey; Yingying Li; Brandon F. Keele; Frederic Bibollet-Ruche; Yuanyuan Guo; Paul A. Goepfert; Mario L. Santiago; Jean Bosco N Ndjango; Cecile Neel; Stephen L. Clifford; Crickette M. Sanz; Shadrack Kamenya; Michael L. Wilson; Anne E. Pusey; Nicole Gross-Camp; Christophe Boesch; Vince Smith; Koichiro Zamma; Michael A. Huffman; John C. Mitani; David P. Watts; Martine Peeters; George M. Shaw; William M. Switzer; Paul M. Sharp; Beatrice H. Hahn

Identifying microbial pathogens with zoonotic potential in wild-living primates can be important to human health, as evidenced by human immunodeficiency viruses types 1 and 2 (HIV-1 and HIV-2) and Ebola virus. Simian foamy viruses (SFVs) are ancient retroviruses that infect Old and New World monkeys and apes. Although not known to cause disease, these viruses are of public health interest because they have the potential to infect humans and thus provide a more general indication of zoonotic exposure risks. Surprisingly, no information exists concerning the prevalence, geographic distribution, and genetic diversity of SFVs in wild-living monkeys and apes. Here, we report the first comprehensive survey of SFVcpz infection in free-ranging chimpanzees (Pan troglodytes) using newly developed, fecal-based assays. Chimpanzee fecal samples (n = 724) were collected at 25 field sites throughout equatorial Africa and tested for SFVcpz-specific antibodies (n = 706) or viral nucleic acids (n = 392). SFVcpz infection was documented at all field sites, with prevalence rates ranging from 44% to 100%. In two habituated communities, adult chimpanzees had significantly higher SFVcpz infection rates than infants and juveniles, indicating predominantly horizontal rather than vertical transmission routes. Some chimpanzees were co-infected with simian immunodeficiency virus (SIVcpz); however, there was no evidence that SFVcpz and SIVcpz were epidemiologically linked. SFVcpz nucleic acids were recovered from 177 fecal samples, all of which contained SFVcpz RNA and not DNA. Phylogenetic analysis of partial gag (616 bp), pol-RT (717 bp), and pol-IN (425 bp) sequences identified a diverse group of viruses, which could be subdivided into four distinct SFVcpz lineages according to their chimpanzee subspecies of origin. Within these lineages, there was evidence of frequent superinfection and viral recombination. One chimpanzee was infected by a foamy virus from a Cercopithecus monkey species, indicating cross-species transmission of SFVs in the wild. These data indicate that SFVcpz (i) is widely distributed among all chimpanzee subspecies; (ii) is shed in fecal samples as viral RNA; (iii) is transmitted predominantly by horizontal routes; (iv) is prone to superinfection and recombination; (v) has co-evolved with its natural host; and (vi) represents a sensitive marker of population structure that may be useful for chimpanzee taxonomy and conservation strategies.


Journal of Virology | 2007

Unique Mutational Patterns in the Envelope α2 Amphipathic Helix and Acquisition of Length in gp120 Hypervariable Domains Are Associated with Resistance to Autologous Neutralization of Subtype C Human Immunodeficiency Virus Type 1

Rong Rong; S. Gnanakaran; Julie M. Decker; Frederic Bibollet-Ruche; Jesse Taylor; Jeffrey N. Sfakianos; John L. Mokili; Mark Muldoon; Joseph Mulenga; Susan Allen; Beatrice H. Hahn; George M. Shaw; Jerry L. Blackwell; Bette Korber; Eric Hunter; Cynthia A. Derdeyn

ABSTRACT Autologous neutralizing antibodies (NAb) against human immunodeficiency virus type 1 generate viral escape variants; however, the mechanisms of escape are not clearly defined. In a previous study, we determined the susceptibilities of 48 donor and 25 recipient envelope (Env) glycoproteins from five subtype C heterosexual transmission pairs to NAb in donor plasma by using a virus pseudotyping assay, thereby providing an ideal setting to probe the determinants of susceptibility to neutralization. In the present study, acquisition of length in the Env gp120 hypervariable domains was shown to correlate with resistance to NAb in donor plasma (P = 0.01; Kendalls tau test) but not in heterologous plasma. Sequence divergence in the gp120 V1-to-V4 region also correlated with resistance to donor (P = 0.0002) and heterologous (P = 0.001) NAb. A mutual information analysis suggested possible associations of nine amino acid positions in V1 to V4 with NAb resistance to the donors antibodies, and five of these were located within an 18-residue amphipathic helix (α2) located on the gp120 outer domain. High nonsynonymous-to-synonymous substitution (dN/dS) ratios, indicative of positive selection, were also found at these five positions in subtype C sequences in the database. Nevertheless, exchange of the entire α2 helix between resistant donor Envs and sensitive recipient Envs did not alter the NAb phenotype. The combined mutual information and dN/dS analyses suggest that unique mutational patterns in α2 and insertions in the V1-to-V4 region are associated with NAb resistance during subtype C infection but that the selected positions within the α2 helix must be linked to still other changes in Env to confer antibody escape. These findings suggest that subtype C viruses utilize mutations in the α2 helix for efficient viral replication and immune avoidance.


Journal of Virology | 2007

Generation of Infectious Molecular Clones of Simian Immunodeficiency Virus from Fecal Consensus Sequences of Wild Chimpanzees

Jun Takehisa; Matthias H. Kraus; Julie M. Decker; Yingying Li; Brandon F. Keele; Frederic Bibollet-Ruche; Kenneth P. Zammit; Zhiping Weng; Mario L. Santiago; Shadrack Kamenya; Michael L. Wilson; Anne E. Pusey; Elizabeth Bailes; Paul M. Sharp; George M. Shaw; Beatrice H. Hahn

ABSTRACT Studies of simian immunodeficiency viruses (SIVs) in their endangered primate hosts are of obvious medical and public health importance, but technically challenging. Although SIV-specific antibodies and nucleic acids have been detected in primate fecal samples, recovery of replication-competent virus from such samples has not been achieved. Here, we report the construction of infectious molecular clones of SIVcpz from fecal viral consensus sequences. Subgenomic fragments comprising a complete provirus were amplified from fecal RNA of three wild-living chimpanzees and sequenced directly. One set of amplicons was concatenated using overlap extension PCR. The resulting clone (TAN1.24) contained intact genes and regulatory regions but was replication defective. It also differed from the fecal consensus sequence by 76 nucleotides. Stepwise elimination of all missense mutations generated several constructs with restored replication potential. The clone that yielded the most infectious virus (TAN1.910) was identical to the consensus sequence in both protein and long terminal repeat sequences. Two additional SIVcpz clones were constructed by direct synthesis of fecal consensus sequences. One of these (TAN3.1) yielded fully infectious virus, while the second one (TAN2.69) required modification at one ambiguous site in the viral pol gene for biological activity. All three reconstructed proviruses produced infectious virions that replicated in human and chimpanzee CD4+ T cells, were CCR5 tropic, and resembled primary human immunodeficiency virus type 1 isolates in their neutralization phenotype. These results provide the first direct evidence that naturally occurring SIVcpz strains already have many of the biological properties required for persistent infection of humans, including CD4 and CCR5 dependence and neutralization resistance. Moreover, they outline a new strategy for obtaining medically important “SIV isolates” that have thus far eluded investigation. Such isolates are needed to identify viral determinants that contribute to cross-species transmission and host adaptation.


Journal of Clinical Investigation | 2012

Efficient SIVcpz replication in human lymphoid tissue requires viral matrix protein adaptation

Frederic Bibollet-Ruche; Anke Heigele; Brandon F. Keele; Juliet L. Easlick; Julie M. Decker; Jun Takehisa; Gerald H. Learn; Paul M. Sharp; Beatrice H. Hahn; Frank Kirchhoff

SIVs infecting wild-living apes in west central Africa have crossed the species barrier to humans on at least four different occasions, one of which spawned the AIDS pandemic. Although the chimpanzee precursor of pandemic HIV-1 strains must have been able to infect humans, the capacity of SIVcpz strains to replicate in human lymphoid tissues (HLTs) is not known. Here, we show that SIVcpz strains from two chimpanzee subspecies are capable of replicating in human tonsillary explant cultures, albeit only at low titers. However, SIVcpz replication in HLT was significantly improved after introduction of a previously identified human-specific adaptation at position 30 in the viral Gag matrix protein. An Arg or Lys at this position significantly increased SIVcpz replication in HLT, while the same mutation reduced viral replication in chimpanzee-derived CD4(+) T cells. Thus, naturally occurring SIVcpz strains are capable of infecting HLTs, the major site of HIV-1 replication in vivo. However, efficient replication requires the acquisition of a host-specific adaptation in the viral matrix protein. These results identify Gag matrix as a major determinant of SIVcpz replication fitness in humans and suggest a critical role in the emergence of HIV/AIDS.


PLOS ONE | 2010

4E10-resistant HIV-1 isolated from four subjects with rare membrane-proximal external region polymorphisms.

Kyle J. Nakamura; Johannes S. Gach; Laura Jones; Katherine Semrau; Jan Walter; Frederic Bibollet-Ruche; Julie M. Decker; Laura Heath; William D. Decker; Chipepo Kankasa; Donald M. Thea; James I. Mullins; Louise Kuhn; Michael B. Zwick; Grace M. Aldrovandi

Human antibody 4E10 targets the highly conserved membrane-proximal external region (MPER) of the HIV-1 transmembrane glycoprotein, gp41, and has extraordinarily broad neutralizing activity. It is considered by many to be a prototype for vaccine development. In this study, we describe four subjects infected with viruses carrying rare MPER polymorphisms associated with resistance to 4E10 neutralization. In one case resistant virus carrying a W680G substitution was transmitted from mother to infant. We used site-directed mutagenesis to demonstrate that the W680G substitution is necessary for conferring the 4E10-resistant phenotype, but that it is not sufficient to transfer the phenotype to a 4E10-sensitive Env. Our third subject carried Envs with a W680R substitution causing variable resistance to 4E10, indicating that residues outside the MPER are required to confer the phenotype. A fourth subject possessed a F673L substitution previously associated with 4E10 resistance. For all three subjects with W680 polymorphisms, we observed additional residues in the MPER that co-varied with position 680 and preserved charged distributions across this region. Our data provide important caveats for vaccine development targeting the MPER. Naturally occurring Env variants described in our study also represent unique tools for probing the structure-function of HIV-1 envelope.


Archive | 2002

The Evolution of Primate Lentiviruses and the Origins of AIDS

Elizabeth Bailes; Roy R. Chaudhuri; Mario L. Santiago; Frederic Bibollet-Ruche; Beatrice H. Hahn; Paul M. Sharp

The acquired immunodeficiency syndrome (AIDS) was first recognized in 1981 (Blattner, 1991) and has become the most globally devastating of the emerging infectious diseases threatening human health in the 21st century. AIDS can be caused by either of the two human immunodeficiency viruses, HlV-1 and HIV-2. It is estimated that so far around 60 million people have been infected by these viruses, of whom 19 million have died of AIDS (UNAIDS, 2001). Combination antiviral therapies have provided clinical relief to some, but these treatments can have toxic sideeffects, and due to their expense these drugs have not yet been widely available in many of the countries worst affected by AIDS. Given the sudden appearance of AIDS in the 20th century, there is an obvious interest in understanding the origins and evolution of the causative viruses, not least in the context of attempts to prevent further such diseases (Hahn et al., 2000).


Retrovirology | 2006

Nef-mediated TCR-CD3 and MHC-I down-modulation prevents CD4+ T cell depletion in natural SIV infection

Michael Schindler; Jan Münch; Olaf Kutsch; Hui Li; Mario L. Santiago; Frederic Bibollet-Ruche; Michaela Müller-Trutwin; Francis J. Novembre; Martine Peeters; Valérie Courgnaud; Elizabeth Bailes; Pierre Roques; Donald L. Sodora; Paul M. Sharp; Guido Silvestri; Beatrice H. Hahn; Frank Kirchhoff

High level immune activation and apoptosis represent a hallmark of HIV-1 infection that is absent from non-pathogenic SIV infections. Recently, we reported that nef alleles from most primate lentiviruses, including HIV-2, down-modulate TCR-CD3 from HIV- or SIV-infected human and sooty mangabey T-cells, thereby blocking their responsiveness to activation (Cell 2006, 125:1055). In contrast, nef alleles from HIV-1 and a subset of closely related SIVs fail to down-regulate TCR-CD3 and to inhibit activation-induced cell death. Thus, differences in Nef function likely provide a mechanism for the varying levels of immune activation observed in pathogenic and non-pathogenic primate lentiviral infections. To further assess the role Nef funtion in vivo we functionally characterized nef alleles derived from 11 SIVsmm-infected mangabeys with >500 CD4+ T-cells/μl and from 15 animals showing a substantial loss of CD4+ T-cells. Our results showed that nef alleles from sooty mangabeys with low CD4+ T cells counts exhibited significantly reduced activity in TCR-CD3 and class I MHC (MHC-I) down-modulation, compared to those derived from animals with normal CD4+ T counts. Thus, our data strongly suggest that the ability of Nef (i) to down-modulate TCR-CD3 and to prevent programmed cell death and (ii) to down-regulate MHC-I to reduce CTL lysis of virally infected CD4+ T cells helps the natural hosts of SIV infection to maintain normal CD4+ T cell counts despite high levels of viral replication.

Collaboration


Dive into the Frederic Bibollet-Ruche's collaboration.

Top Co-Authors

Avatar

Beatrice H. Hahn

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

George M. Shaw

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martine Peeters

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julie M. Decker

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Mario L. Santiago

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Yingying Li

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jun Takehisa

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge